EP4135851A1 - Gegen flt3 gerichtete chimäre antigenrezeptormodifizierte zellen zur behandlung von flt3-positiven malignomen - Google Patents

Gegen flt3 gerichtete chimäre antigenrezeptormodifizierte zellen zur behandlung von flt3-positiven malignomen

Info

Publication number
EP4135851A1
EP4135851A1 EP21724442.5A EP21724442A EP4135851A1 EP 4135851 A1 EP4135851 A1 EP 4135851A1 EP 21724442 A EP21724442 A EP 21724442A EP 4135851 A1 EP4135851 A1 EP 4135851A1
Authority
EP
European Patent Office
Prior art keywords
cell
cells
seq
nucleic acid
acid molecule
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP21724442.5A
Other languages
English (en)
French (fr)
Inventor
Michael A. Caligiuri
Jianhua Yu
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
City of Hope
Original Assignee
City of Hope
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by City of Hope filed Critical City of Hope
Publication of EP4135851A1 publication Critical patent/EP4135851A1/de
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4613Natural-killer cells [NK or NK-T]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464454Enzymes
    • A61K39/464462Kinases, e.g. Raf or Src
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70596Molecules with a "CD"-designation not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5156Animal cells expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5158Antigen-pulsed cells, e.g. T-cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/48Blood cells, e.g. leukemia or lymphoma
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16041Use of virus, viral particle or viral elements as a vector
    • C12N2740/16043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • This disclosure concerns chimeric antigen receptors (CAR) engineered to bind to FMS-like tyrosine kinase 3 (FLT3) expressing cells, T cells or NK cells expressing such CAR, methods of formulating such CAR T or NK cells and methods of use as anti-cancer agents.
  • CAR chimeric antigen receptors
  • FMS-like tyrosine kinase 3 is a transmembrane protein expressed on normal hematopoietic stem and progenitor cells. It is also expressed on malignant blasts in acute myeloid leukemia (AML).
  • Cells expressing a FLT3 -specific chimeric antigen receptor can suppress in vivo growth of solid and blood cancers such as leukemia, and prolong the survival of leukemia-bearing mice. (Chen et al., Leukemia 31:1830, 2017). Thus, remains needed is a FLT3-specific CAR therapy that efficiently treats a cancer in a patient.
  • FLT3-CAR T or NK cells could be considered for treatment of refractory disease or up front treatment, for example in the elderly with AML, given their dismal prognosis with standard therapy.
  • nucleic acid molecules encoding a FLT3 -specific CAR
  • the nucleic acid molecules include an improved coding sequence (e.g., SEQ ID NO: 1) for a FLT3 -specific CAR that allows higher expression of the CAR and greater effectiveness than a previously designed coding sequence.
  • the FLT3 CAR nucleic acid molecule comprises, or consists essentially of, or yet further consists of: ATGGGGTGGTCAAGCATTATTCTGTTTCTGGTCGCTACCGCTACAGGCGTCCATC
  • CATGCAGGCCCTGCCCCCTCGC (SEQ ID NO: 1).
  • SEQ ID NO: 1 encodes a polypeptide having the sequence:
  • the CAR includes, comprises, or consists essentially of, or yet further consists of: a signal sequence (e.g., MGWSSIILFLVATATGVH; SEQ ID NO:3); a FLT3 -targeted single chain variable fragment (scFv)
  • a signal sequence e.g., MGWSSIILFLVATATGVH; SEQ ID NO:3
  • scFv FLT3 -targeted single chain variable fragment
  • LEPK S CDKTHT CPPCPDPKGT SEQ ID NO: 5
  • a CD28 transmembrane domain F WVL VVV GGVL AC Y SLL VT VAFIIF W V ; SEQ ID NO: 6
  • a CD28 co-stimulatory domain RKRSRLLHSD YMNMTPRRPGPTRKHY QP Y APPRDF AAYRS ; SEQ ID NO:7
  • the scFv comprises, or consists essentially of, or yet further consists of a heavy chain variable region:
  • a peptide linker e.g
  • the CAR can be produced using a vector in which the CAR open reading frame is followed by a T2A ribosome skip sequence and a truncated EGFR (referred to herein as EGFRt or tEGFR) or truncated CD 19 (referred to herein as CD19t or tCD19).
  • EGFRt or tEGFR truncated EGFR
  • CD19t or tCD19 tCD19
  • the EGFRt or CD19t incorporated in a vector such as a lentiviral or retroviral vector can act as suicide gene to ablate the CAR+ T or NK cells in cases of treatment-related toxicity.
  • nucleic acid molecule encoding an anti- FMS-like tyrosine kinase 3 (FLT3) chimeric antigen receptor (CAR).
  • the nucleic acid molecule comprises, or alternatively consists essentially of, or yet further consists of the polynucleotide sequence of nucleotide (nt) 55 to nt 777 of SEQ ID NO:l or an equivalent thereof that encodes the FLT3 scFv antigen binding fragment.
  • the equivalent is at least about 75% (including but not limited to at least about 90%, or at least about 95%, or at least about 99%) identical to nucleotide (nt) 55 to nt 777 of SEQ ID NO: 1 (that encodes the FLT3 scFv antigen binding fragment) while maintaining at least one of the optimized nucleotide changes. Additionally or alternatively, the equivalent comprises, or alternatively consists essentially of, or yet further consists of the polynucleotide of nucleotide (nt) 55 to nt 777 of SEQ ID NO: 12 or an equivalent thereof that encodes the FLT3 scFv antigen binding fragment.
  • the equivalent does not comprise, or consist essentially of, or consist of (nt) 55 to nt 777 of SEQ ID NO: 14.
  • the nucleic acid molecule further comprises, or consists essentially of, or yet further consists of a polynucleotide encoding a truncated CD 19 or a truncated EGFR.
  • the nucleic acid molecule comprises, or alternatively consists essentially of, or yet further consists of the polynucleotide sequence of nucleotide (nt) 1 to nt 777 of SEQ ID NO: 1 or an equivalent thereof that encodes a signal peptide and the FLT3 scFv antigen binding region.
  • the equivalent is at least about 75% (including but not limited to at least about 90%, or at least about 95%, or at least about 99%) identical to nucleotide (nt) 1 to nt 777 of SEQ ID NO: 1 while maintaining the optimized nucleotide changes.
  • the equivalent comprises, or alternatively consists essentially of, or yet further consists of the polynucleotide of nucleotide (nt) 1 to nt 777 of SEQ ID NO: 12. In further embodiments, the equivalent does not comprise, or consist essentially of, or consist of (nt) 1 to nt 777 of SEQ ID NO: 14.
  • the nucleic acid molecule comprises, or alternatively consists essentially of, or yet further consists of the polynucleotide sequence of SEQ ID NO: 1 or an equivalent thereof.
  • the equivalent is at least about 75% (including but not limited to at least about 90%, or at least about 95%, or at least about 99%) identical to SEQ ID NO: 1 while maintaining the optimized nucleotide changes.
  • the equivalent comprises, or alternatively consists essentially of, or yet further consists of the polynucleotide of SEQ ID NO: 12.
  • a vector comprising, or alternatively consisting essentially of, or yet further consisting of a nucleic acid molecule encoding the FLT3 CAR as disclosed herein or a complementary nucleic acid molecule thereof.
  • the vector is a plasmid vector, a viral vector, an expression vector.
  • the vector is a retroviral vector.
  • the vector is a lentiviral vector.
  • a population of human T or NK cells comprising a nucleic acid molecule as disclosed herein encoding the FLT3 CAR or a vector as disclosed herein encoding the FLT3 CAR.
  • an isolated cell comprising a nucleic acid molecule as disclosed herein encoding the FLT3 CAR or a vector as disclosed herein encoding the FLT3 CAR, or a population thereof.
  • the cell is selected from the group of: an immune cell, an NK cell, a T cell, a stem cell, a progenitor cell or a precursor cell to the immune cell, the NK cell or the T cell.
  • composition comprising, or alternatively consisting essentially of, or yet further consisting of a population of cells containing or expressing the FLT3 CAR as disclosed herein, or an isolated cell expressing the FLT3 CAR as disclosed herein, and a carrier, and optionally a stabilizer, preservative or cryopreservative.
  • a method of treating a human patient suffering from cancer such as acute myeloid leukemia.
  • the method comprises, or alternatively consists essentially of, or yet further consists of administering a population of autologous or allogeneic human T or NK cells comprising a nucleic acid molecule encoding the FLT3 CAR as disclosed herein or a vector comprising the nucleic acid molecule encoding the FLT3 CAR as disclosed herein.
  • a reduction in tumor burden such as a reduction in tumor burden, inhibition of metastasis, enhanced overall survival, reduced toxicity, prolonged progression free survival, slowing or halting of cancer, reduction in tumor markers or other clinical symptoms as compared to treatment with other known therapies or in the absence of therapy.
  • These methods can be combined with other known anti-tumor or cancer therapies as a second line, third line, fourth line, or fifth line therapy, or be provided as a first line therapy.
  • a method of treating a patient in need thereof comprises, or alternatively consists essentially of, or yet further consists of administering a FLT3 CAR cell population as disclosed herein to the patient.
  • a method of preparing CAR T or NK cells comprises, or alternatively consists essentially of, or yet further consists of providing a population of autologous or allogeneic human T or NK cells and transducing the T or NK cells with a vector as disclosed herein or a nucleic acid molecule as disclosed herein.
  • the method comprises, or alternatively consists essentially of, or yet further consists of transducing the cell with a vector as disclosed herein or a nucleic acid molecule as disclosed herein.
  • compositions and kits for use in a method as disclosed herein is also provided.
  • FIGURE 1 provides a schematic diagram showing how pHIV-7 was constructed.
  • FIGURE 2 provides an illustration of pHIV-7.
  • FIGURE 3 provides an illustration of a plasmid for producing a retroviral vector genome expressing the FLT3 -specific CAR.
  • FIGURES 4A - 4D provide flow cytometry results evaluating CAR expression on four T cell groups, i.e., T cells expressing GFP serving as a control (FIGURE 4A), T cell expressing tCD19 also serving as a control (FIGURE 4B), T cells expressing tCD19 and FLT3 -specific CAR encoded by the nucleic acid molecule without optimization (FIGURE 4C), and T cells expressing tCD19 and FLT3-specific CAR encoded by the optimized nucleic acid molecule (FIGURE 4D).
  • T cells expressing GFP serving as a control
  • T cell expressing tCD19 also serving as a control
  • FIGURE 4C T cells expressing tCD19 and FLT3 -specific CAR encoded by the nucleic acid molecule without optimization
  • FIGURE 4D T cells expressing tCD19 and FLT3-specific CAR encoded by the optimized nucleic acid molecule
  • FIGURES 5A - 5B plot tumor lysis percentages tested at two different effector to target cell ratios (E:T) using two different target cancer cells, i.e., MOLM13 and U937.
  • FIGURE 5A shows results tested at an E:T of 1 :5, while FIGURE 5B shows results tested at an E:T of 1 :25.
  • T cells expressing GFP GFP
  • T cells expressing tCD19 and FLT3-specific CAR encoded by the optimized nucleic acid molecule After Opt
  • tumor lysis percentages were plotted as bars in a set of four following the same order.
  • FIGURES 6A - 6C provide concentrations of interleukin 2 (IL-2) in the supernatant of co-culture of cancer cells and T cells isolated from Donor 1 to Donor 3, respectively.
  • the T cells were engineered to express GFP (GFP control), or tCD19 (tCD19 control), or tCD19 and FLT3 -specific CAR encoded by the nucleic acid molecule without optimization (Before Opt. FLT3/CD19), or tCD19 and FLT3-specific CAR encoded by the optimized nucleic acid molecule (After Opt. FLT3/CD19).
  • MOLM13 cells are FLT3 expressing (i.e., FLT3+) tumor target while U937 cells do not express FLT3 and served as a negative control. Accordingly, two bars are plotted in FIGURES 6A-6C for each Donor and each T cell group: the left one indicates IL-2 concentration in the co-culture with MOLM13, while the right one indicates IL-2 concentration in the co-culture with U937.
  • FIGURES 7A - 7C provide concentrations of Interferon gamma (i.e., IFNy or IFN-gamma) in the supernatant of co-culture of cancer cells and T cells isolated from Donor 1 to Donor 3, respectively.
  • the T cells were engineered to express GFP (GFP control), or tCD19 (tCD19 control), or tCD19 and FLT3 -specific CAR encoded by the nucleic acid molecule without optimization (Before Opt. FLT3/CD19), or tCD19 and FLT3-specific CAR encoded by the optimized nucleic acid molecule (After Opt. FLT3/CD19). And then each group of the engineered T cells was incubated with two cancer cell lines separately:
  • MOLM13 cells are FLT3+ tumor target while U937 cells do not express FLT3 and served as a negative control. Accordingly, two bars are plotted in FIGURES 7A-7C for each Donor and each T cell group: the left one indicates IFNy concentration in the co-culture with MOLM13, while the right one indicates IFNy concentration in the co-culture with U937.
  • FIGURE 8 shows that FLT3-specific CAR cells slow/halt AML progression in dose-dependent manner.
  • FLT3-specific CAR expressing NK cells were compared with FLT3-specific CAR expressing T cells in a MOLM-13 model of FLT3(+) AML. All work was performed with viably frozen primary human CAR NK cells derived from cord blood. DETAILED DESCRIPTION
  • a cell includes a plurality of cells, including mixtures thereof.
  • compositions and methods are intended to mean that the compounds, compositions and methods include the recited elements, but not exclude others.
  • Consisting essentially of when used to define compounds, compositions and methods, shall mean excluding other elements of any essential significance to the combination. Thus, a composition consisting essentially of the elements as defined herein would not exclude trace contaminants, e.g., from the isolation and purification method and pharmaceutically acceptable carriers, preservatives, and the like. “Consisting of’ shall mean excluding more than trace elements of other ingredients. Embodiments defined by each of these transition terms are within the scope of this technology.
  • comparative terms as used herein can refer to certain variation from the reference.
  • such variation can refer to about 10%, or about 20%, or about 30%, or about 40%, or about 50%, or about 60%, or about 70%, or about 80%, or about 90%, or about 1 fold, or about 2 folds, or about 3 folds, or about 4 folds, or about 5 folds, or about 6 folds, or about 7 folds, or about 8 folds, or about 9 folds, or about 10 folds, or about 20 folds, or about 30 folds, or about 40 folds, or about 50 folds, or about 60 folds, or about 70 folds, or about 80 folds, or about 90 folds, or about 100 folds or more higher than the reference.
  • such variation can refer to about 1%, or about 2%, or about 3%, or about 4%, or about 5%, or about 6%, or about 7%, or about 8%, or about 0%, or about 10%, or about 20%, or about 30%, or about 40%, or about 50%, or about 60%, or about 70%, or about 75%, or about 80%, or about 85%, or about 90%, or about 95%, or about 96%, or about 97%, or about 98%, or about 99% of the reference.
  • substantially or “essentially” means nearly totally or completely, for instance, 95% or greater of some given quantity. In some embodiments, “substantially” or “essentially” means 95%, 96%, 97%, 98%, 99%, 99.5%, or 99.9%.
  • animal refers to living multi-cellular vertebrate organisms, a category that includes, for example, mammals and birds.
  • mammal includes both human and non-human mammals.
  • a mammal is a human.
  • mammals include humans, non-human primates (e.g., apes, gibbons, chimpanzees, orangutans, monkeys, macaques, and the like), domestic animals (e.g., dogs and cats), farm animals (e.g., horses, cows, goats, sheep, pigs) and experimental animals (e.g., mouse, rat, rabbit, guinea pig).
  • a mammal is a human.
  • a mammal can be any age or at any stage of development (e.g., an adult, teen, child, infant, or a mammal in utero).
  • a mammal can be male or female.
  • a subject is a human.
  • a subject has or is diagnosed of having or is suspected of having a disease.
  • the terms “first” “second” “third” “fourth” or similar in a component name are used to distinguish and identify more than one components sharing certain identity in their names. For example, “first administration” and “second administration” are used across the specification to distinguishing two administrations.
  • polynucleotide “nucleic acid” and “nucleic acid molecule” are used interchangeably and refer to a polymeric form of nucleotides of any length, either deoxyribonucleotides or ribonucleotides or analogs thereof. Polynucleotides can have any three-dimensional structure and may perform any function, known or unknown.
  • polynucleotides a gene or gene fragment (for example, a probe, primer, EST or SAGE tag), exons, introns, messenger RNA (mRNA), transfer RNA, ribosomal RNA, RNAi, ribozymes, cDNA, recombinant polynucleotides, branched polynucleotides, plasmids, vectors, isolated DNA of any sequence, isolated RNA of any sequence, nucleic acid probes and primers.
  • a polynucleotide can comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs. If present, modifications to the nucleotide structure can be imparted before or after assembly of the polynucleotide.
  • sequence of nucleotides can be interrupted by non-nucleotide components.
  • a polynucleotide can be further modified after polymerization, such as by conjugation with a labeling component.
  • the term also refers to both double- and single-stranded molecules. Unless otherwise specified or required, any aspect of this technology that is a polynucleotide encompasses both the double-stranded form and each of two complementary single-stranded forms known or predicted to make up the double-stranded form.
  • this term includes, but is not limited to, single-, double-, or multi-stranded DNA or RNA, genomic DNA, cDNA, DNA-RNA hybrids, or a polymer comprising purine and pyrimidine bases or other natural, chemically or biochemically modified, non-natural, or derivatized nucleotide bases.
  • an equivalent to a reference nucleic acid, polynucleotide or oligonucleotide encodes the same sequence encoded by the reference.
  • an equivalent to a reference nucleic acid, polynucleotide or oligonucleotide hybridizes to the reference, a complement reference, a reverse reference, and/or a reverse-complement (i.e., complementary) reference, optionally under conditions of high stringency.
  • an equivalent nucleic acid, polynucleotide or oligonucleotide is one having at least 70%, or at least 75%, or at least 80 % sequence identity, or alternatively at least 85 % sequence identity, or alternatively at least 90 % sequence identity, or alternatively at least 92 % sequence identity, or alternatively at least 95 % sequence identity, or alternatively at least 97 % sequence identity, or alternatively at least 98 % sequence identity to the reference nucleic acid, polynucleotide, or oligonucleotide, or alternatively an equivalent nucleic acid hybridizes under conditions of high stringency to a reference polynucleotide or its complement.
  • an equivalent has at least the 70%, or at least 75%, or at least 80 % sequence identity, or alternatively at least 85 % sequence identity, or alternatively at least 90 % sequence identity, or alternatively at least 92 % sequence identity, or alternatively at least 95 % sequence identity, or alternatively at least 97 % sequence identity, or alternatively at least 98 % sequence identity to the reference nucleic acid, polynucleotide, or oligonucleotide, or alternatively an equivalent nucleic acid hybridizes under conditions of high stringency to a reference polynucleotide or its complement.
  • Hybridization refers to a reaction in which one or more polynucleotides react to form a complex that is stabilized via hydrogen bonding between the bases of the nucleotide residues.
  • the hydrogen bonding may occur by Watson-Crick base pairing, Hoogstein binding, or in any other sequence-specific manner.
  • the complex may comprise two strands forming a duplex structure, three or more strands forming a multi-stranded complex, a single self-hybridizing strand, or any combination of these.
  • a hybridization reaction may constitute a step in a more extensive process, such as the initiation of a PCR reaction, or the enzymatic cleavage of a polynucleotide by a ribozyme.
  • Examples of stringent hybridization conditions include: incubation temperatures of about 25°C to about 37°C; hybridization buffer concentrations of about 6x SSC to about 10x SSC; formamide concentrations of about 0% to about 25%; and wash solutions from about 4x SSC to about 8x SSC.
  • Examples of moderate hybridization conditions include: incubation temperatures of about 40°C to about 50°C; buffer concentrations of about 9x SSC to about 2x SSC; formamide concentrations of about 30% to about 50%; and wash solutions of about 5x SSC to about 2x SSC.
  • high stringency conditions include: incubation temperatures of about 55°C to about 68°C; buffer concentrations of about lx SSC to about 0.1x SSC; formamide concentrations of about 55% to about 75%; and wash solutions of about lx SSC, 0.1x SSC, or deionized water.
  • hybridization incubation times are from 5 minutes to 24 hours, with 1, 2, or more washing steps, and wash incubation times are about 1, 2, or 15 minutes.
  • SSC is 0.15 MNaCl and 15 mM citrate buffer. It is understood that equivalents of SSC using other buffer systems can be employed.
  • a coding nucleic acid molecule can be optimized, for example, to have a certain GC content.
  • GC-content is the percentage of nitrogenous bases in a nucleic acid molecule that are either guanine (G) or cytosine (C). This measure indicates the proportion of G and C bases (i.e., G and C residues) out of an implied four total bases, also including adenine and thymine in DNA and adenine and uracil in RNA. Quantitatively, each GC base pair is held together by three hydrogen bonds, while AT and AU base pairs are held together by two hydrogen bonds. Accordingly, a nucleic acid molecule with low GC-content has a lower thermos-stability compared to that with high GC-content.
  • codon refers to a sequence of three consecutive nucleotides that corresponds with a specific amino acid residue or a stop signal during protein synthesis.
  • the codon is a standard genetic code as provided in the table below.
  • codon frequency of a nucleic acid molecule refers to the rate of recurrence of a codon used by the nucleic acid molecule upon expressing a polypeptide.
  • a codon frequency as used herein is presented as the percentage of the number of the codon over the total codon number of the full nucleic acid molecule.
  • a codon frequency as used herein is presented as the average number of the codon per the 1000 codons.
  • a nucleic acid molecular having 300 nucleotide residues uses 100 codons, 10 of which are a codon consisting of GCC and encoding an alanine (Ala) amino acid residue.
  • the GCC codon frequency may be presented as 10%, or 100 per 1000 codons.
  • Several online tools are available for calculating codon frequency of a nucleic acid, such as www.bioinformatics.org/sms2/codon_usage.
  • encode refers to a polynucleotide which is said to “encode” a polypeptide if, in its native state or when manipulated by methods well known to those skilled in the art, can be transcribed and/or translated to produce the mRNA for the polypeptide and/or a fragment thereof.
  • the antisense strand is the complement of such a nucleic acid, and the encoding sequence can be deduced therefrom.
  • a “gene” refers to a polynucleotide containing at least one open reading frame (ORF) that is capable of encoding a particular polypeptide or protein after being transcribed and translated.
  • ORF open reading frame
  • the term “express” refers to the production of a gene product.
  • the term “expression” refers to the process by which polynucleotides are transcribed into mRNA or the process by which the transcribed mRNA is subsequently being translated into peptides, polypeptides, or proteins, or both processes. If the polynucleotide is derived from genomic DNA, expression may include splicing of the mRNA in a eukaryotic cell. The expression level of a gene may be determined by measuring the amount of mRNA or protein in a cell or tissue sample.
  • transduce or “transduction” as it is applied to the production of CAR expressing cells refers to the process whereby a foreign nucleotide sequence is introduced into a cell. In some embodiments, this transduction is done via a vector.
  • the term “vector” refers to a nucleic acid construct deigned for transfer between different hosts, including but not limited to a plasmid, a virus, a cosmid, a phage, a BAC, a YAC, etc.
  • plasmid vectors may be prepared from commercially available vectors.
  • viral vectors may be produced from baculoviruses, retroviruses, adenoviruses, AAVs, etc. according to techniques known in the art.
  • the viral vector is a lentiviral vector.
  • the viral vector is a retroviral vector.
  • a “plasmid” is an extra-chromosomal DNA molecule separate from the chromosomal DNA which is capable of replicating independently of the chromosomal DNA. In many cases, it is circular and double-stranded. Plasmids provide a mechanism for horizontal gene transfer within a population of microbes and typically provide a selective advantage under a given environmental state. Plasmids may carry genes that provide resistance to naturally occurring antibiotics in a competitive environmental niche, or alternatively the proteins produced may act as toxins under similar circumstances. Many plasmids are commercially available for such uses.
  • the gene to be replicated is inserted into copies of a plasmid containing genes that make cells resistant to particular antibiotics and a multiple cloning site (MCS, or polylinker), which is a short region containing several commonly used restriction sites allowing the easy insertion of DNA fragments at this location.
  • MCS multiple cloning site
  • one or more plasmids are used in producing a viral vector or a viral genome.
  • a plasmid is used for replicating or amplifying a polynucleotide.
  • Another major use of plasmids is to make large amounts of proteins. In this case, researchers grow bacteria containing a plasmid harboring the gene of interest. Just as the bacterium produces proteins to confer its antibiotic resistance, it can also be induced to produce large amounts of proteins from the inserted gene. This is a cheap and easy way of mass-producing a gene or the protein it then codes for.
  • a “viral vector” is defined as a recombinantly produced virus or viral particle that comprises a polynucleotide to be delivered into a host cell, either in vivo, ex vivo or in vitro.
  • viral vectors include retroviral vectors, lentiviral vectors, adenovirus vectors, adeno-associated virus vectors, alphavirus vectors and the like.
  • Alphavirus vectors such as Semliki Forest virus-based vectors and Sindbis virus-based vectors, have also been developed for use in gene therapy and immunotherapy. See, Schlesinger and Dubensky (1999) Curr. Opin. Biotechnol. 5:434-439 and Ying, et al. (1999) Nat. Med. 5(7):823-827.
  • a vector construct refers to the polynucleotide comprising the lentiviral genome or part thereof, and a therapeutic gene.
  • lentiviral mediated gene transfer or “lentiviral transduction” carries the same meaning and refers to the process by which a gene or nucleic acid sequences are stably transferred into the host cell by virtue of the virus entering the cell and integrating its genome into the host cell genome.
  • the virus can enter the host cell via its normal mechanism of infection or be modified such that it binds to a different host cell surface receptor or ligand to enter the cell.
  • Retroviruses carry their genetic information in the form of RNA; however, once the virus infects a cell, the RNA is reverse-transcribed into the DNA form which integrates into the genomic DNA of the infected cell.
  • the integrated DNA form is called a provirus.
  • lentiviral vector refers to a viral particle capable of introducing exogenous nucleic acid into a cell through a viral or viral-like entry mechanism.
  • lentiviral vector is a type of retroviral vector well-known in the art that has certain advantages in transducing non-dividing cells as compared to other retroviral vectors. See, Trono D. (2002) Lentiviral vectors, New York: Spring-Verlag Berlin Heidelberg.
  • Lentiviral vectors of this disclosure are based on or derived from oncoretroviruses (the sub-group of retroviruses containing MLV), and lentiviruses (the sub group of retroviruses containing HIV). Examples include ASLV, SNV and RSV all of which have been split into packaging and vector components for lentiviral vector particle production systems.
  • the lentiviral vector particle according to the disclosure may be based on a genetically or otherwise (e.g. by specific choice of packaging cell system) altered version of a particular retrovirus.
  • That the vector particle according to the disclosure is "based on” or “derived from” a particular virus means that the vector is derived from that particular virus.
  • the genome of the vector particle comprises components from that virus as a backbone.
  • the virus is a retrovirus.
  • the vector particle contains essential vector components compatible with the RNA genome, including reverse transcription and integration systems. Usually these will include gag and pol proteins derived from the particular retrovirus. Thus, the majority of the structural components of the vector particle will normally be derived from that retrovirus, although they may have been altered genetically or otherwise so as to provide desired useful properties. However, certain structural components and in particular the env proteins, may originate from a different virus.
  • the vector host range and cell types infected or transduced can be altered by using different env genes in the vector particle production system to give the vector particle a different specificity.
  • AAV adeno-associated virus
  • AAV adeno-associated virus
  • AAV refers to a member of the class of viruses associated with this name and belonging to the genus dependoparvovirus, family Parvoviridae. Multiple serotypes of this virus are known to be suitable for gene delivery; all known serotypes can infect cells from various tissue types. At least 11 sequentially numbered, AAV serotypes are known in the art.
  • Non-limiting exemplary serotypes useful in the methods disclosed herein include any of the 11 serotypes, e.g., AAV2, AAV8, AAV9, or variant or synthetic serotypes, e.g., AAV-DJ and AAV PHP.B.
  • the AAV particle comprises, alternatively consists essentially of, or yet further consists of three major viral proteins: VP1, VP2 and VP3.
  • the AAV refers to of the serotype AAV1, AAV2, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, AAV 12, AAV13, AAV PHP.B, or AAV rh74. These vectors are commercially available or have been described in the patent or technical literature.
  • a regulatory sequence intends a polynucleotide that is operatively linked to a target polynucleotide to be transcribed and/or replicated, and facilitates the expression and/or replication of the target polynucleotide.
  • the term “operatively linked” refers to an association between the regulatory polynucleotide and the polynucleotide sequence to which it is linked such that, when a specific protein binds to the regulatory polynucleotide, the linked polynucleotide is transcribed.
  • a promoter is an example of an expression control element or a regulatory sequence.
  • the term “promoter” as used herein refers to any sequence that regulates the expression of a coding sequence. Promoters can be located 5’ or upstream of a gene or other polynucleotide, that provides a control point for regulated gene transcription. Polymerase II and III are examples of promoters. Promoters may be constitutive, inducible, repressible, or tissue-specific, for example.
  • a “promoter” is a control sequence that is a region of a polynucleotide sequence at which initiation and rate of transcription are controlled. It may contain genetic elements at which regulatory proteins and molecules may bind such as RNA polymerase and other transcription factors. Non-limiting examples of promoters include an EF1 alpha promoter, a Cytomegalovirus (CMV) promoter, and an MMLV promoter.
  • CMV Cytomegalovirus
  • EFlalpha also referred to herein as EF-lalpha
  • EF-lalpha promoter sequence is known in the art (see, e.g., addgene.org/11154/sequences/; ncbi.nlm.nih.gov/nuccore/J04617, each last accessed on March 13, 2019, and Zheng and Baum (2014) IntT. J. Med. Sci.
  • an EFlalpha promoter comprises, or consists essentially of, or yet further consists of AAGGATCTGCGATCGCTCCGGTGCCCGTCAGTGGGCAGAGCGCACATCGCCCAC
  • CMV cytomegalovirus
  • an CMV promoter comprises, or consists essentially of, or yet further consists of atcgattggctcatgtccaacattaccgccatgttgacattgattattgactagttattaatagtaatcaattacggggtcattagttcatagc ccatatatggagttccgcgttacataacttacggtaaatggcccgcctggctgaccgcccaacgacccccgcccattgacgtcaataat gacgtatgttcccatagtaacgccaatagggactttccattgacgtcaatgggtggagtattttacggtaaactgcccacttggcagtagtcaatgggtggagtattttacggtaaactgcccacttggcagtacat caagtgtatcatatgccaagtacgcc
  • an EF1 alpha promoter comprises, or consists essentially of, or yet further consists of SEQ ID NO: 24.
  • the term “enhancer”, as used herein, denotes sequence elements that augment, improve or ameliorate transcription of a nucleic acid sequence irrespective of its location and orientation in relation to the nucleic acid sequence to be expressed.
  • An enhancer may enhance transcription from a single promoter or simultaneously from more than one promoter. As long as this functionality of improving transcription is retained or substantially retained (e.g., at least 70%, at least 80%, at least 90% or at least 95% of wild-type activity, that is, activity of a full-length sequence), any truncated, mutated or otherwise modified variants of a wild-type enhancer sequence are also within the above definition.
  • protein refers to a compound of two or more subunit amino acids, amino acid analogs or peptidomimetics.
  • the subunits may be linked by peptide bonds. In another aspect, the subunit may be linked by other bonds, e.g., ester, ether, etc.
  • a protein or peptide must contain at least two amino acid residues and no limitation is placed on the maximum number of amino acids which may comprise a protein’s or peptide’s sequence.
  • amino acid refers to either natural and/or unnatural or synthetic amino acids, including glycine and both the D and L optical isomers, amino acid analogs and peptidomimetics.
  • an equivalent protein or polypeptide is one having at least about 60%, at least about 70%, at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, or at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity to the reference protein or polypeptide.
  • an equivalent protein or polypeptide has at least about 60%, at least about 70%, at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, or at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity to a polypeptide or protein as disclosed herein.
  • an equivalent protein or polypeptide has at least about 60%, at least about 70%, at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, or at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity to polypeptide or protein encoded by an equivalent polynucleotide as noted herein.
  • the equivalent of a polynucleotide would encode a protein or polypeptide of the same or similar function as the reference or parent polynucleotide.
  • the equivalent is a functional protein that optionally can be identified through one or more assays described herein or otherwise available to one of skill in the art.
  • an equivalent has at least about 60%, at least about 70%, at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, or at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity to the reference protein or polypeptide.
  • an amino acid (aa) or nucleotide (nt) residue position in a sequence of interest “corresponding to” or “aligned to” an identified position in a reference sequence refers to that the residue position is aligned to the identified position in a sequence alignment between the sequence of interest and the reference sequence.
  • Various programs are available for performing such sequence alignments, such as Clustal Omega and BLAST.
  • the term “specifically recognizing and binding” means the contact between an antibody (or an antigen binding fragment thereof, or a biological agent comprising such antibody or antigen binding fragment, such as a CAR or a CAR expressing cell) and its target antigen with a binding affinity substantially higher than the binding affinity with another molecule.
  • the binding affinity between the antibody (or an antigen binding fragment thereof, or a biological agent comprising such antibody or antigen binding fragment, such as a CAR or a CAR expressing cell) and its target antigen is at least KG 3 M, including any number or any range therein, such as at least about 10 -6 M, or at least about 10 -7 M, and preferably 10 -8 M, 10 -9 M, 10 -10 M, 10 -11 M, or 10 -12 M.
  • FLT3 and “FMS-like tyrosine kinase 3” are used interchangeably, referring to a receptor-type tyrosine-protein kinase FLT3 associated with this name, any of its alternate names (Fms-Related Tyrosine Kinase, Stem Cell Tyrosine Kinase, Fms-Like Tyrosine Kinase, FL Cytokine Receptor, CD135 Antigen, EC 2.7.10.1, CD135, FLK-2, STK1, FLK2, Growth Factor Receptor Tyrosine Kinase Type III, Receptor- Type Tyrosine-Protein Kinase FLT3, Fetal Liver Kinase 2, Fetal Liver Kinase-2, EC 2.7.10 , FLT-3, or STK-1) or UniProt Accession No.
  • Non-limiting examples of FLT3 include: Human FLT3 Isoform 1 consisting of
  • an activating mutation in Fms-like tyrosine kinase 3 refers to a mutant FLT3 nucleotide sequence or amino acid sequence leading to an activation of the FLT3 kinase, such as a FLT3 internal tandem duplication (ITD) mutation.
  • FLT3 is mutated in about one third of acute myeloid leukemia cases.
  • the most frequent FLT3 mutations in acute myeloid leukemia are internal tandem duplication (ITD) mutations in the juxtamembrane domain (23%) and point mutations in the tyrosine kinase domain (10%).
  • the most frequent kinase domain mutation is the substitution of aspartic acid at position 838 (equivalent to the human aspartic acid residue at position 835) with a tyrosine (FLT3/D835Y), converting aspartic acid to tyrosine.
  • FLT3 tyrosine
  • FLT3/D835Y tyrosine
  • chimeric antigen receptor refers to a fused protein comprising an extracellular domain capable of binding to an antigen, a transmembrane domain derived from a polypeptide different from a polypeptide from which the extracellular domain is derived, and at least one intracellular domain.
  • the “chimeric antigen receptor (CAR)” is sometimes called a “chimeric receptor”, a “T-body”, or a “chimeric immune receptor (CIR).”
  • extracellular domain capable of binding to an antigen means any oligopeptide or polypeptide that can bind to a certain antigen, such as an antibody or an antigen binding fragment thereof.
  • antibody also referred to herein as “immunoglobulin” collectively refers to immunoglobulins or immunoglobulin-like molecules including by way of example and without limitation, IgA, IgD, IgE, IgG and IgM, combinations thereof, and similar molecules produced during an immune response in any vertebrate, for example, in mammals such as humans, goats, rabbits and mice, as well as nonmammalian species, such as shark immunoglobulins.
  • the term “antibody” includes intact immunoglobulins and “antibody fragments” or “antigen binding fragments” that specifically bind to a molecule of interest (or a group of highly similar molecules of interest) to the substantial exclusion of binding to other molecules (for example, antibodies and antibody fragments that have a binding constant for the molecule of interest that is at least 10 3 M -1 or greater, at least 10 4 M -1 or greater or at least 10 5 M -1 or greater than a binding constant for other molecules in a biological sample).
  • the term “antibody” also includes genetically engineered forms such as chimeric antibodies (for example, murine or humanized non-primate antibodies), or heteroconjugate antibodies (such as, bispecific antibodies), or both. See also, Pierce Catalog and Handbook, 1994-1995 (Pierce Chemical Co., Rockford, Ill.); Owen et al., Kuby Immunology, 7 th Ed., W.H.
  • an immunoglobulin has heavy (H) chains and light (L) chains interconnected by disulfide bonds.
  • Each heavy and light chain contains a constant region and a variable region, (the regions are also known as "domains").
  • the heavy and the light chain variable regions specifically bind the antigen.
  • Light and heavy chain variable regions contain a "framework" region interrupted by three hypervariable regions, also called “complementarity-determining regions" or "CDRs".
  • framework region and CDRs have been defined (see, Kabat et al., Sequences of Proteins of Immunological Interest, U.S. Department of Health and Human Services, 1991, which is hereby incorporated by reference).
  • the Kabat database is now maintained online.
  • the sequences of the framework regions of different light or heavy chains are relatively conserved within a species.
  • the framework region of an antibody that is the combined framework regions of the constituent light and heavy chains, largely adopts a b-sheet conformation and the CDRs form loops which connect, and in some cases form part of, the b-sheet structure.
  • framework regions act to form a scaffold that provides for positioning the CDRs in correct orientation by inter-chain, non-covalent interactions.
  • the CDRs are primarily responsible for binding to an epitope of an antigen.
  • the CDRs of each chain are typically referred to as CDR1, CDR2, and CDR3, numbered sequentially starting from the N-terminus, and are also typically identified by the chain in which the particular CDR is located (heavy chain regions labeled CDRH and light chain regions labeled CDRL).
  • CDRH3 is the CDR3 from the variable domain of the heavy chain of the antibody in which it is found
  • a CDRLl is the CDR1 from the variable domain of the light chain of the antibody in which it is found.
  • an FLT3 antibody will have a specific VH region and the VL region sequence unique to the FLT3 relevant antigen, and thus specific CDR sequences.
  • Antibodies with different specificities i.e., different combining sites for different antigens
  • have different CDRs Although it is the CDRs that vary from antibody to antibody, only a limited number of amino acid positions within the CDRs are directly involved in antigen binding. These positions within the CDRs are called specificity determining residues (SDRs).
  • the term “antigen” refers to a compound, composition, or substance that may be specifically bound by the products of specific humoral or cellular immunity, such as an antibody molecule or T-cell receptor.
  • Antigens can be any type of molecule including, for example, haptens, simple intermediary metabolites, sugars (e.g., oligosaccharides), lipids, and hormones as well as macromolecules such as complex carbohydrates (e.g., polysaccharides), phospholipids, and proteins.
  • antigens include, but are not limited to, viral antigens, bacterial antigens, fungal antigens, protozoa and other parasitic antigens, tumor antigens, antigens involved in autoimmune disease, allergy and graft rejection, toxins, and other miscellaneous antigens.
  • antigen of a binding moiety such as an antibody, an antigen binding fragment thereof, or a CAR
  • a binding moiety such as an FLT3 CAR or an FLT3 -specific CAR
  • antigen such as an antigen and the binding moiety after the antigen (such as an anti-FLT3 antibody)
  • the binding moiety followed by “to” or “directed to” and then the antigen (such as an antibody to FLT3).
  • the term “antigen binding domain” refers to any protein or polypeptide domain that can specifically bind to an antigen target, such as FLT3.
  • the antigen binding fragment may be selected from the group consisting of Fab, F(ab’)2, Fab’, scFv, or Fv.
  • a single chain Fv fragment comprises, or consists essentially of, or yet further consists of a heavy chain variable region and a light chain variable region connected with a linker peptide (typically around 5 to 25 amino acids in length).
  • the variable regions of the heavy chain and the light chain may be derived from the same antibody or different antibodies.
  • an FLT3 scFv is disclosed herein as SEQ ID NO: 4. Additional suitable FLT3 scFv can be found, for example, in US Patent No. 10,961,312 and W02020/010284, each of which is incorporated herein by reference in its entirety.
  • linker sequence “linker peptide” and “linker polypeptide” are used interchangeably, relating to any amino acid sequence comprising from 1 to 10, or alternatively, 8 amino acids, or alternatively 6 amino acids, or alternatively 5 amino acids that may be repeated from 1 to 10, or alternatively to about 8, or alternatively to about 6, or alternatively about 5, or 4 or alternatively 3, or alternatively 2 times.
  • the linker may comprise up to 15 amino acid residues consisting of a pentapeptide repeated three times.
  • the linker sequence is a (Glycine4Serine)3 (aa 119 to aa 133 of SEQ ID NO: 4) flexible polypeptide linker comprising three copies of Gly-Gly-Gly-Gly-Ser (SEQ ID NO: 27).
  • the terms “linker sequence” “linker peptide” and “linker polypeptide” refer to a peptide consists of some (for example, about 1 to about 50) random amino acid residues.
  • the term “equivalent” or “biological equivalent” of an antibody means the ability of the antibody to selectively bind its epitope/antigen protein or fragment thereof as measured by ELISA or other suitable methods.
  • Biologically equivalent antibodies include, but are not limited to, those antibodies, peptides, antibody fragments, antibody variant, antibody derivative and antibody mimetics that bind to the same epitope as the reference antibody.
  • the “transmembrane domain” means any oligopeptide or polypeptide known to span the cell membrane and that can function to link the extracellular and signaling domains.
  • the transmembrane domain may be derived either from a natural or from a synthetic source. Where the source is natural, the domain may be derived from any membrane-bound or transmembrane protein.
  • Transmembrane regions of particular use in this disclosure may be derived from CD8, CD28, CD3, CD45, CD4, CD5, CDS, CD9, CD 16, CD22, CD33, CD37, CD64, CD80, CD86, CD 134, CD137, CD 154, and TCR.
  • the transmembrane domain may be synthetic, in which case it will comprise predominantly hydrophobic residues such as leucine and valine. Preferably a triplet of phenylalanine, tryptophan and valine will be found at each end of a synthetic transmembrane domain.
  • a short oligo- or polypeptide linker preferably between 2 and 10 amino acids in length may form the linkage between the transmembrane domain and the cytoplasmic signaling domain of the CAR.
  • a glycine-serine doublet provides a particularly suitable linker.
  • the transmembrane domain comprises, or alternatively consists essentially of, or yet consists of a CD8 a transmembrane domain or a CD28 transmembrane domain.
  • CD28 transmembrane domain refers to a specific protein fragment associated with this name and any other molecules that have analogous biological function that share at least 70%, or alternatively at least 80% amino acid sequence identity, at least 90% sequence identity, or alternatively at least 95% sequence identity with the CD28 transmembrane domain sequence as shown herein.
  • GenBank Accession Nos: XM_006712862.2 and XM_009444056.1 provide additional, non-limiting, example sequences of the CD28 transmembrane domain.
  • the sequences associated with each of the listed accession numbers are incorporated herein by reference in its entirety.
  • the CD28 transmembrane domain comprises, or alternatively consists essentially of, or yet consists of SEQ ID NO: 6 or an equivalent thereof.
  • the equivalent of SEQ ID NO: 6 may comprises 1, or 2, or 3, or 4, or 5, or 6, or 7, or 8, or 9, or 10, or 11, or 12, or 13, or 14, or 15, or more mutations compared to SEQ ID NO: 6 but is still capable of spanning the cell membrane and functioning to link the extracellular and signaling domains as SEQ ID NO: 6.
  • intracellular domain means any oligopeptide or polypeptide known to function as a domain that transmits a signal to cause activation or inhibition of a biological process in a cell.
  • the intracellular signaling domain of the CAR is responsible for activation of at least one of the traditional effector functions of an immune cell in which a CAR has been placed.
  • the intracellular signaling domain refers to a portion of a protein which transduces the effector function signal and directs the immune cell to perform its specific function. An entire signaling domain or a truncated portion thereof may be used so long as the truncated portion is sufficient to transduce the effector function signal.
  • Cytoplasmic sequences of the TCR and co-receptors as well as derivatives or variants thereof can function as intracellular signaling domains for use in a CAR.
  • Intracellular signaling domains of particular use in this disclosure may be derived from FcR, TCR, CD3, CDS, CD22, CD79a, CD79b, and CD66d.
  • the intracellular signaling domain of the CAR can comprise, or alternatively consist essentially of, or yet consist of a CD3 ⁇ (i.e., CD3zeta) signaling domain.
  • CD3 zeta signaling domain or “CD3 zeta intracellular signaling domain” refers to a specific protein fragment associated with this name and any other molecules that have analogous biological function that share at least 70%, or alternatively at least 80% amino acid sequence identity, preferably 90% sequence identity, more preferably at least 95% sequence identity with the CD3 zeta signaling domain sequence as shown herein.
  • the CD3 zeta signaling domain comprises, or alternatively consists essentially of, or yet consists of
  • the equivalent of SEQ ID NO: 8 may comprises 1, or 2, or 3, or 4, or 5, or 6, or 7, or 8, or 9, or 10, or 11, or 12, or 13, or 14, or 15, or more mutations compared to SEQ ID NO: 8 but is still capable of transducing the effector function signal and directing the immune cell to perform its specific function as SEQ ID NO: 8. Exemplified methods assessing such transduction can be found, for example, in Bridgeman JS, et al. Clin Exp Immunol. 2014 Feb;175(2):258-67.
  • a secondary or co-stimulatory signal may also be required.
  • a co-stimulatory signaling molecule also referred to herein as a costimulatory domain or a costimulatory signaling domain or a costimulatory signaling region
  • the intracellular region of a co-stimulatory signaling molecule including but not limited the intracellular domains of the proteins CD27, CD28, 4- IBB (CD 137), 0X40, CD30, CD40, PD-1, ICOS (CD278), lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, NKG2D, DAPIO, DAP 12, 2B3, 4B2, B7- H3, MHC class I molecule, TNF receptor proteins, Immunoglobulin-like proteins, cytokine receptors, integrins, signaling lymphocytic activation molecules (SLAM proteins), activating NK cell receptors, BTLA, a To
  • the intracellular domain may comprise, alternatively consist essentially of, or yet further comprise one or more costimulatory signaling domains in addition to the primary signaling domain.
  • a CAR may comprise one, two, or more costimulatory domains, in addition to a signaling domain (e.g., a CD3 z signaling domain).
  • the intracellular domain further comprises one or more or two or more costimulatory regions selected from a CD28 costimulatory signaling region, a 4- IBB costimulatory signaling region, an ICOS costimulatory signaling region, or an 0X40 costimulatory region.
  • the cell activation moiety (e.g., the cytoplasmic region) of the chimeric antigen receptor is a T cell or an NK cell signaling domain comprising, or alternatively consisting essentially of, or yet further consisting of, one or more proteins or fragments thereof selected from the group consisting of CD8 protein, CD28 protein, 4-1BB protein, 0X40, CD30, CD40, PD-1, ICOS, LFA-1, CD2, CD7, CD27, LIGHT, NKG2C, B7- H3, and CD3-zeta protein.
  • the cell activation moiety (e.g., the cytoplasmic region) of the chimeric antigen receptor is a T cell or an NK cell signaling domain comprising, or alternatively consisting essentially of, or yet further consisting of, one or more proteins or fragments thereof selected from the group consisting of CD8 protein, CD28 protein, 4-1BB protein, and CD3zeta protein.
  • CD28 costimulatory signaling region or a “CD28 costimulatory domain” refers to a specific protein fragment associated with this name and any other molecules that have analogous biological function that share at least 70%, or alternatively at least 80% amino acid sequence identity, preferably 90% sequence identity, more preferably at least 95% sequence identity with the CD28 costimulatory signaling region sequence shown herein.
  • the example sequences CD28 costimulatory signaling domain are provided in U.S. Patent No. 5,686,281; Geiger, T.L. et al., Blood 98: 2364-2371 (2001); Hombach, A. et al., J Immunol 167: 6123-6131 (2001); Maher, J.
  • the CD28 costimulatory domain comprises, or alternatively consists essentially of, or yet consists of SEQ ID NO: 7 or an equivalent thereof.
  • the equivalent of SEQ ID NO: 7 may comprises 1, or 2, or 3, or 4, or 5, or 6, or 7, or 8, or 9, or 10, or 11, or 12, or 13, or 14, or 15, or more mutations compared to SEQ ID NO: 7 but is still capable of stimulating the immune cell to perform its specific function as SEQ ID NO: 7.
  • a signal peptide (sometimes referred to as signal sequence, targeting signal, localization signal, localization sequence, transit peptide, leader sequence or leader peptide) is a short peptide (usually 16-30 amino acids long) present at the N-terminus of the majority of newly synthesized proteins that are destined toward the secretory pathway. These synthesized proteins are the directed to reside either inside certain organelles (the endoplasmic reticulum, Golgi or endosomes), secreted from the cell, or inserted into most cellular membranes. In some embodiments, a signal peptide as disclosed herein direct the protein to be expressed on the cell membrane.
  • a chimeric antigen receptor may optionally comprise a “hinge domain” which serves as a linker between the extracellular and transmembrane domains.
  • a spacer domain (also referred to herein as a spacer or a hinge or a hinge domain) is an extracellular structural region of the CAR that separates the binding units from the transmembrane domain. These spacers generally supply stability for efficient CAR expression and activity.
  • Such a domain may comprise, or consist essentially of, or yet further consist of, for example, a portion of a human Fc domain, a CH3 domain, or the hinge region of any immunoglobulin, such as IgA, IgD, IgE, IgG, or IgM, or variants thereof.
  • some embodiments may comprise an IgG4 hinge with or without a S228P, L235E, and/or N297Q mutation (according to Kabat numbering). Additional spacers include, but are not limited to, CD4, CD8, and CD28 hinge regions.
  • the hinge domain may be derived either from a natural or from a synthetic source. In some embodiments, the hinge domain is derived from a cluster of differentiation protein, such as CD8, CD28, CD3, CD45, CD4, CD5, CDS, CD9, CD 16, CD22, CD33, CD37, CD64, CD80, CD86, CD 134, CD137, CD 154. In one embodiments, the hinge domain is a CD8 a hinge domain.
  • the hinge domain is derived from an immunoglobulin, such as IgA, IgD, IgE, IgG, or IgM.
  • the hinge domain is an IgGl hinge domain.
  • the IgGl hinge domain comprises, or alternatively consists essentially of, or yet consists of LEPK S CDKTHT CPPCPDPKGT (SEQ ID NO: 5) or an equivalent thereof.
  • an equivalent of SEQ ID NO: 5 comprises 1, or 2, or 3, or 4, or 5, or 6, or 7, or 8, or 9, or 10, or 11, or 12, or 13, or 14, or 15, or more mutations compared to SEQ ID NO: 5 but is still capable of substantially maintaining the stability for efficient CAR expression and activity as SEQ ID NO: 5.
  • a protein expressed on cell surface may be used as a marker or to provide a suicide switch of a CAR expressing cell as disclosed herein.
  • a portion of or the whole cytoplasmic region of such protein is usually truncated so that the native function of the protein is reduced or even abolished.
  • a protein is referred to herein as a truncated protein marker.
  • the truncated protein marker when used as a suicide switch of the CAR expressing cell, does not express or is expressed at a substantially lower level on a normal cell or a normal cell adjacent to the CAR expressing cell in the subject.
  • a normal cell of the subject upon removal of the CAR expressing cell (for example, by administering a neutralizing antibody specially recognizing and binding the truncated protein marker, or by administering a toxin conjugated to a moiety directing the toxin to the truncated protein marker,), a normal cell of the subject would not be jeopardized.
  • the disclosure herein shows that certain truncated protein markers (for example, a truncated CD 19, or a truncated EGFR) are expressed at a higher rate compared to other markers (such as a green fluorescent protein (GFP)). See, FIGURE 4B versus. FIGURE 4A.
  • those truncated protein markers are particular suitable for using in a CAR expressing cell as disclosed herein.
  • CD 19 truncated protein markers
  • B-lymphocyte antigen CD 19 are used interchangeably to refer to a protein known to be a transmembrane protein that in humans is encoded by the gene CD 19. In humans, CD 19 is expressed in all B lineage cells, except for plasma cells, and in follicular dendritic cells.
  • Non-limiting exemplary sequences of this protein or the underlying gene may be found under Gene Cards ID: GC16P028943, HGNC: 1633, Entrez Gene: 930, Ensembl: ENSG00000177455, OMIM: 107265, and UniProtKB: P15391, which are incorporated by reference herein.
  • An exemplified truncated CD 19 is provided herein and comprising, or consisting essentially of, or yet further consisting of MPPPRLLFFLLFLTPMEVRPEEPLVVKVEEGDNAVLQCLKGTSDGPTQQLTWSRESP LKPFLKLSLGLPGLGIHMRPLAIWLFIFNVSQQMGGFYLCQPGPPSEKAWQPGWTVN VEGSGELFRWNV SDLGGLGCGLKNRS SEGPS SPSGKLMSPKL YVWAKDRPEIWEGE PPCVPPRDSLNQSLSQDLTMAPGSTLWLSCGVPPDSVSRGPLSWTHVHPKGPKSLLS LELKDDRP ARDMW VMET GLLLPR AT AQD AGK Y Y CHRGNLTM SFHLEIT ARP VLWH WLLRTGGWK V S AVTL AYLIF CLC SL VGILHLQRALVLRRKR (SEQ ID NO: 13).
  • an equivalent of SEQ ID NO: 13 or 11, for example those comprising 1, or 2, or 3, or 4, or 5, or 6, or 7, or 8, or 9, or 10, or 11, or 12, or 13, or 14, or 15, or more mutations compared to SEQ ID NO: 13 or 11 or those having at least 90%, at least 95%, at least 98% identical to SEQ ID NO: 13 or 11, may be used.
  • an equivalent of SEQ ID NO: 13 or 11 is still capable of being recognized and bound to by a moiety, such as an antibody or an antigen binding fragment thereof, specifically recognizing and binding CD19.
  • the equivalent of SEQ ID NO: 13 or 11 does not direct a cell expressing the equivalent to perform a function as a wild type CD 19 does.
  • the polynucleotide encoding SEQ ID NO: 11 comprises or consists essentially of, or yet further consists of
  • EGFR truncated protein marker
  • EGFR Epidermal Growth Factor Receptor
  • HGNC HGNC
  • NCBI Entrez Gene 1956
  • OMIM® 131550
  • UniProtKB/Swiss-Prot P00533, each of which is incorporated by reference herein by its entirety.
  • An exemplified truncated EGFR is provided herein and comprising, or consisting essentially of, or yet further consisting of L VT SLLLCELPHP AFLLIPRK V CN GIGIGEFKD SL SIN ATNIKHFKNCT SI S GDLHILP V A FRGDSFTHTPPLDPQELDILKTVKEITGFLLIQAWPENRTDLHAFENLEIIRGRTKQHG QFSLAVVSLNITSLGLRSLKEISDGDVIISGNKNLCYANTINWKKLFGTSGQKTKIISN RGEN S CK AT GQ V CH ALC SPEGC W GPEPRDC V S CRN V SRGREC VDKCNLLEGEPREF VENSECIQCHPECLPQAMNITCTGRGPDNCIQCAHYIDGPHCVKTCPAGVMGENNTL VWK Y AD AGH V CHL CHPN C T Y GC T GP GLEGCPTN GPKIP S I AT GM V G ALLLLL V V AL GIGL
  • an equivalent of SEQ ID NO: 10 for example those comprising 1, or 2, or 3, or 4, or 5, or 6, or 7, or 8, or 9, or 10, or 11, or 12, or 13, or 14, or 15, or more mutations compared to SEQ ID NO: 10 or those having at least 90%, at least 95%, at least 98% identical to SEQ ID NO: 10, may be used.
  • an equivalent of SEQ ID NO: 10 is still capable of being recognized and bound to by a moiety, such as an antibody or an antigen binding fragment thereof, specifically recognizing and binding EGFR.
  • the equivalent of SEQ ID NO: 10 does not direct a cell expressing the equivalent to perform a function as a wild type EGFR does.
  • a ribosomal skip sequence which is also referred to as a cleavable peptide, or a cleavable linker, means a peptide that can be cleaved, for example, by an enzyme or a peptide that can induce ribosomal skipping during translation of a protein in a cell, or both.
  • One translated polypeptide comprising such cleavable peptide can produce two final products, therefore, allowing expressing more than one polypeptides from one open reading frame.
  • a ribosomal skip sequence can be used to express a CAR and a truncated protein marker on a cell as disclosed herein.
  • nucleic acid molecule comprising a CAR coding sequence, a truncated protein marker coding sequence, an internal ribosome entry site (IRES) located therebetween.
  • cleavable peptides is a self-cleaving peptide, such as a 2A self-cleaving peptide.
  • 2A self-cleaving peptides is a class of 18-22 aa-long peptides, which can induce the cleaving of the recombinant protein in a cell.
  • the 2 A self-cleaving peptide is selected from P2A, T2A, E2A, F2A and BmCPV2A. See, for example, Wang Y, et al. 2A self-cleaving peptide-based multi-gene expression system in the silkworm Bombyx mori. Sci Rep. 2015;5: 16273. Published 2015 Nov 5.
  • T2A and 2A peptide are used interchangeably to refer to any 2A peptide or fragment thereof, any 2A-like peptide or fragment thereof, or an artificial peptide comprising the requisite amino acids in a relatively short peptide sequence (on the order of 20 amino acids long depending on the virus of origin) containing the consensus polypeptide motif D-V/I-E-X-N-P-G-P (SEQ ID NO: 29), wherein X refers to any amino acid generally thought to be self-cleaving.
  • IRES internal ribosome entry site
  • mRNA messenger RNA
  • IRES also refers to a polynucleotide sequence (such as an RNA sequence, a DNA sequence or a hybrid thereof) complementary, or reverse, or both complementary and reverse to an IRES RNA sequence.
  • Non-limiting examples of IRES can be found in Hellen CU and Samow P. Internal ribosome entry sites in eukaryotic mRNA molecules. Genes Dev. 2001 Jul 1 ; 15(13): 1593-612.
  • Detectable label “label”, “detectable marker” or “marker” are used interchangeably, including, but not limited to radioisotopes, fluorochromes, chemiluminescent compounds, dyes, and proteins, including enzymes. Detectable labels can also be attached to a polynucleotide, polypeptide, antibody or composition described herein.
  • label or a detectable label intends a directly or indirectly detectable compound or composition that is conjugated directly or indirectly to the composition to be detected, e.g., N-terminal histidine tags (N-His), magnetically active isotopes, e.g., 115 Sn, 117 Sn and 119 Sn, a non-radioactive isotopes such as 13 C and 15 N, polynucleotide or protein such as an antibody so as to generate a “labeled” composition.
  • N-terminal histidine tags N-His
  • magnetically active isotopes e.g., 115 Sn, 117 Sn and 119 Sn
  • a non-radioactive isotopes such as 13 C and 15 N
  • polynucleotide or protein such as an antibody so as to generate a “labeled” composition.
  • the term also includes sequences conjugated to the polynucleotide that will provide a signal upon expression of the inserted sequence
  • the label may be detectable by itself (e.g., radioisotope labels or fluorescent labels) or, in the case of an enzymatic label, may catalyze chemical alteration of a substrate compound or composition which is detectable.
  • the labels can be suitable for small scale detection or more suitable for high-throughput screening.
  • suitable labels include, but are not limited to magnetically active isotopes, non-radioactive isotopes, radioisotopes, fluorochromes, chemiluminescent compounds, dyes, and proteins, including enzymes.
  • the label may be simply detected, or it may be quantified.
  • a response that is simply detected generally comprises a response whose existence merely is confirmed
  • a response that is quantified generally comprises a response having a quantifiable (e.g., numerically reportable) value such as an intensity, polarization, or other property.
  • the detectable response may be generated directly using a luminophore or fluorophore associated with an assay component actually involved in binding, or indirectly using a luminophore or fluorophore associated with another (e.g., reporter or indicator) component.
  • luminescent labels that produce signals include, but are not limited to bioluminescence and chemiluminescence.
  • Detectable luminescence response generally comprises a change in, or an occurrence of a luminescence signal.
  • Suitable methods and luminophores for luminescently labeling assay components are known in the art and described for example in Haugland, Richard P. (1996) Handbook of Fluorescent Probes and Research Chemicals (6th ed).
  • Examples of luminescent probes include, but are not limited to, aequorin and luciferases.
  • the term “immunoconjugate” comprises an antibody or an antibody derivative associated with or linked to a second agent, such as a cytotoxic agent, a detectable agent, a radioactive agent, a targeting agent, a human antibody, a humanized antibody, a chimeric antibody, a synthetic antibody, a semisynthetic antibody, or a multispecific antibody.
  • a second agent such as a cytotoxic agent, a detectable agent, a radioactive agent, a targeting agent, a human antibody, a humanized antibody, a chimeric antibody, a synthetic antibody, a semisynthetic antibody, or a multispecific antibody.
  • fluorescent labels include, but are not limited to, fluorescein, rhodamine, tetramethylrhodamine, eosin, erythrosin, coumarin, methyl- coumarins, pyrene, Malacite green, stilbene, Lucifer Yellow, CASCADE BLUETM, and Texas Red.
  • fluorescein fluorescein
  • rhodamine tetramethylrhodamine
  • eosin erythrosin
  • coumarin methyl- coumarins
  • pyrene Malacite green
  • stilbene Lucifer Yellow
  • CASCADE BLUETM Lucifer Yellow
  • Texas Red Texas Red
  • the fluorescent label is functionalized to facilitate covalent attachment to a cellular component present in or on the surface of the cell or tissue such as a cell surface marker.
  • Suitable functional groups include, but are not limited to, isothiocyanate groups, amino groups, haloacetyl groups, maleimides, succinimidyl esters, and sulfonyl halides, all of which may be used to attach the fluorescent label to a second molecule.
  • the choice of the functional group of the fluorescent label will depend on the site of attachment to either a linker, the agent, the marker, or the second labeling agent.
  • a purification label or maker refers to a label that may be used in purifying the molecule or component that the label is conjugated to, such as an epitope tag (including but not limited to a Myc tag, a human influenza hemagglutinin (HA) tag, a FLAG tag), an affinity tag (including but not limited to a glutathione-S transferase (GST), a poly- Histidine (His) tag, Calmodulin Binding Protein (CBP), or Maltose-binding protein (MBP)), or a fluorescent tag.
  • an epitope tag including but not limited to a Myc tag, a human influenza hemagglutinin (HA) tag, a FLAG tag
  • an affinity tag including but not limited to a glutathione-S transferase (GST), a poly- Histidine (His) tag, Calmodulin Binding Protein (CBP), or Maltose-binding protein (MBP)
  • fluorescent tag including but not limited to
  • polypeptide or equivalents of each thereof can be followed by an additional 50 amino acids, or alternatively about 40 amino acids, or alternatively about 30 amino acids, or alternatively about 20 amino acids, or alternatively about 10 amino acids, or alternatively about 5 amino acids, or alternatively about 4, or 3, or 2 or 1 amino acids at the carboxy -terminus.
  • each CAR component as exemplified herein include other proteins that have analogous biological function that share at least 70%, or alternatively at least 80% amino acid sequence identity, preferably 90% sequence identity, more preferably at least 95% sequence identity with the exemplified component.
  • an equivalent intends at least about 70% homology or identity, or at least 80 % homology or identity and alternatively, or at least about 85 %, or alternatively at least about 90 %, or alternatively at least about 95 %, or alternatively 98 % percent homology or identity and exhibits substantially equivalent biological activity to the reference protein, polypeptide or nucleic acid.
  • an equivalent thereof is a polynucleotide that hybridizes under stringent conditions to the reference polynucleotide or its complement.
  • a polynucleotide or polynucleotide region (or a polypeptide or polypeptide region) having a certain percentage (for example, 80%, 85%, 90%, or 95%) of “sequence identity” to another sequence means that, when aligned, that percentage of bases (or residues) are the same in comparing the two sequences.
  • the alignment and the percent homology or sequence identity can be determined using software programs known in the art, for example those described in Current Protocols in Molecular Biology (Ausubel et al., eds. 1987) Supplement 30, section 7.7.18, Table 7.7.1.
  • default parameters are used for alignment.
  • a preferred alignment program is BLAST, using default parameters.
  • Another preferred alignment program is Clustal Omega accessible at www.ebi.ac.uk/Tools/msa/clustalo/ or a Pairwise Sequence Alignment accessible at www.ebi.ac.uk/Tools/psa/, using default parameters.
  • a cell may be a prokaryotic or a eukaryotic cell.
  • the cell is an immune cell.
  • “Host cell” refers not only to the particular subject cell but to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
  • the term “culturing” refers to the in vitro or ex vivo propagation of cells or organisms on or in media of various kinds. It is understood that the descendants of a cell grown in culture may not be completely identical (i.e., morphologically, genetically, or phenotypically) to the parent cell.
  • Immuno cells includes, e.g., white blood cells (leukocytes, such as granulocytes (neutrophils, eosinophils, and basophils), monocytes, and lymphocytes (T cells, B cells, natural killer (NK) cells and NKT cells)), lymphocytes (T cells, B cells, natural killer (NK) cells) and myeloid-derived cells (neutrophil, eosinophil, basophil, monocyte, macrophage, dendritic cells).
  • the immune cell is derived from hematopoietic stem cells (HSCs).
  • the HSCs are produced in the bone marrow.
  • the HSCs are isolated from cord blood.
  • the HSCs are isolated from peripheral blood.
  • the immune cell is derived from one or more of the following: progenitor cells, embryonic stem cells, embryonic stem cell derived cells, embryonic germ cells, embryonic germ cell derived cells, stem cells, stem cell derived cells, pluripotent stem cells, induced pluripotent stem cells (iPSc), hematopoietic stem cells (HSCs), or immortalized cells.
  • the HSC are derived from umbilical cord blood of a subject, peripheral blood of a subject, or bone marrow of a subject.
  • immune cells refer to central memory T cells, NK cells, native memory T cells, pan T cells, or any combination thereof. In some embodiments, immune cells refer to peripheral blood mononuclear cells (PBMCs) substantially depleted for CD25+ cells and CD14+ cells.
  • PBMCs peripheral blood mononuclear cells
  • T cell refers to a type of lymphocyte that matures in the thymus. T cells play an important role in cell-mediated immunity and are distinguished from other lymphocytes, such as B cells, by the presence of a T-cell receptor on the cell surface. T-cells may either be isolated or obtained from a commercially available source. “T cell” includes all types of immune cells expressing CD3 including T-helper cells (CD4+ cells), cytotoxic T-cells (CD8+ cells), natural killer T-cells, T-regulatory cells (Treg) and gamma-delta T cells.
  • CD4+ cells T-helper cells
  • CD8+ cells cytotoxic T-cells
  • Reg T-regulatory cells
  • gamma-delta T cells gamma-delta T cells.
  • a “cytotoxic cell” includes CD8+ T cells, natural-killer (NK) cells, and neutrophils, which cells are capable of mediating cytotoxicity responses.
  • T-cell lines include lines BCL2 (AAA) Jurkat (ATCC® CRL-2902TM), BCL2 (S70A) Jurkat (ATCC® CRL-2900TM), BCL2 (S87A) Jurkat (ATCC® CRL-2901TM), BCL2 Jurkat (ATCC® CRL-2899TM), Neo Jurkat (ATCC® CRL-2898TM), TALL-104 cytotoxic human T cell line (ATCC # CRL-11386).
  • T-cell lines e.g., such as Deglis, EBT-8, HPB-MLp-W, HUT 78, HUT 102, Karpas 384, Ki 225, My-La, Se-Ax, SKW-3, SMZ-1 and T34; and immature T- cell lines, e g., ALL-SIL, Be13, CCRF-CEM, CML-T1, DND-41, DU.528, EU-9, HD- Mar, HPB-ALL, H-SB2, HT-1, JK-T1, Jurkat, Karpas 45, KE-37, KOPT-K1, K-Tl, L-KAW, Loucy, MAT, MOLT-1, MOLT 3, MOLT-4, MOLT 13, MOLT- 16, MT-1, MT-ALL,
  • mature T-cell lines e.g., such as Deglis, EBT-8, HPB-MLp-W, HUT 78, HUT 102, Karpas 384, Ki 225, My-La,
  • HuT78 ATCC CRM-TIB-161
  • MJ[G11] ATCC CRL-8294
  • HuT102 ATCC TIB-162
  • Null leukemia cell lines including but not limited to REH, NALL-1, KM-3, L92-221, are a another commercially available source of immune cells, as are cell lines derived from other leukemias and lymphomas, such as K562 erythroleukemia, THP-1 monocytic leukemia,
  • Non-limiting exemplary sources for such commercially available cell lines include the American Type Culture Collection, or ATCC, (www.atcc.org/) and the German Collection of Microorganisms and Cell Cultures (www.dsmz.de/).
  • NK cell also known as natural killer cell, refers to a type of lymphocyte that originates in the bone marrow and play a critical role in the innate immune system. NK cells provide rapid immune responses against viral-infected cells, tumor cells or other stressed cell, even in the absence of antibodies and major histocompatibility complex on the cell surfaces. NK cells may either be isolated or obtained from a commercially available source.
  • Non-limiting examples of commercial NK cell lines include lines NK-92 (ATCC® CRL-2407TM), NK-92MI (ATCC® CRL-2408TM). Further examples include but are not limited to NK lines HANK1, KHYG-1, NKL, NK-YS, NOI-90, and YT.
  • Non-limiting exemplary sources for such commercially available cell lines include the American Type Culture Collection, or ATCC, (www.atcc.org/) and the German Collection of Microorganisms and Cell Cultures (www.dsmz.de/).
  • stem cell refers to a cell that is in an undifferentiated or partially differentiated state and has the capacity for self-renewal or to generate differentiated progeny or both.
  • Self-renewal is defined as the capability of a stem cell to proliferate and give rise to more such stem cells, while maintaining its developmental potential (i.e., totipotent, pluripotent, multipotent, etc.).
  • embryonic stem cell is used herein to refer to any stem cell derived from non-embryonic tissue, including fetal, juvenile, and adult tissue.
  • Natural somatic stem cells have been isolated from a wide variety of adult tissues including blood, bone marrow, brain, olfactory epithelium, skin, pancreas, skeletal muscle, and cardiac muscle.
  • exemplary naturally occurring somatic stem cells include, but are not limited to, mesenchymal stem cells (MSCs) and neural or neuronal stem cells (NSCs).
  • the stem or progenitor cells can be embryonic stem cells or an induced pluripotent stem cell (iPSC).
  • the stem or progenitor cells are hematopoietic stem cells (HSCs).
  • embryonic stem cells refers to stem cells derived from tissue formed after fertilization but before the end of gestation, including pre- embryonic tissue (such as, for example, a blastocyst), embryonic tissue, or fetal tissue taken any time during gestation, typically but not necessarily before approximately 10-12 weeks gestation. Most frequently, embryonic stem cells are pluripotent cells derived from the early embryo or blastocyst. Embryonic stem cells can be obtained directly from suitable tissue, including, but not limited to human tissue, or from established embryonic cell lines. “Embryonic-like stem cells” refer to cells that share one or more, but not all characteristics, of an embryonic stem cell.
  • “Differentiation” describes the process whereby an unspecialized cell acquires the features of a specialized cell such as a heart, liver, immune or muscle cell.
  • “Directed differentiation” refers to the manipulation of stem cell culture conditions to induce differentiation into a particular cell type.
  • “Dedifferentiated” defines a cell that reverts to a less committed position within the lineage of a cell.
  • the term “differentiates or differentiated” defines a cell that takes on a more committed (“differentiated”) position within the lineage of a cell.
  • the term “differentiates or differentiated” defines a cell that takes on a more committed (“differentiated”) position within the lineage of a cell. “Dedifferentiated” defines a cell that reverts to a less committed position within the lineage of a cell. Induced pluripotent stem cells are examples of dedifferentiated cells.
  • the "lineage" of a cell defines the heredity of the cell, i.e. its predecessors and progeny.
  • the lineage of a cell places the cell within a hereditary scheme of development and differentiation.
  • a “multi-lineage stem cell” or “multipotent stem cell” refers to a stem cell that reproduces itself and at least two further differentiated progeny cells from distinct developmental lineages.
  • the lineages can be from the same germ layer (i.e. mesoderm, ectoderm or endoderm), or from different germ layers.
  • a “precursor” or “precursor cell” or “progenitor cell” intends to mean cells that have a capacity to differentiate into a specific type of cell.
  • a progenitor cell may be a stem cell.
  • a progenitor cell may also be more specific than a stem cell.
  • a progenitor cell may be unipotent or multipotent. Compared to adult stem cells, a progenitor cell may be in a later stage of cell differentiation.
  • An example of progenitor cell includes, without limitation, a progenitor nerve cell.
  • a “pluripotent cell” defines a less differentiated cell that can give rise to at least two distinct (genotypically or phenotypically or both) further differentiated progeny cells.
  • a “pluripotent cell” includes an Induced Pluripotent Stem Cell (iPSC) which is an artificially derived stem cell from a non-pluripotent cell, typically an adult somatic cell, that has historically been produced by inducing expression of one or more stem cell specific genes.
  • iPSC Induced Pluripotent Stem Cell
  • stem cell specific genes include, but are not limited to, the family of octamer transcription factors, i.e.
  • Oct-3/4 the family of Sox genes, i.e., Soxl, Sox2, Sox3, Sox 15 and Sox 18; the family of Klf genes, i.e. Klfl, Klf2, Klf4 and Klf5; the family of Myc genes, i.e. c-myc and L-myc; the family of Nanog genes, i.e., OCT4, NANOG and REX1; or LIN28.
  • Examples of iPSCs are described in Takahashi et al. (2007) Cell advance online publication 20 November 2007; Takahashi & Yamanaka (2006) Cell 126:663-76; Okita et al. (2007) Nature 448:260-262; Yu et al. (2007) Science advance online publication 20 November 2007; and Nakagawa et al. (2007) Nat. Biotechnol. Advance online publication 30 November 2007.
  • An “induced pluripotent cell” intends embryonic-like cells reprogrammed to the immature phenotype from adult cells.
  • Various methods are known in the art, e.g., "A simple new way to induce pluripotency: Acid.” Nature, 29 January 2014 and available at sciencedaily.com/releases/2014/01/140129184445, last accessed on February 5, 2014 and U.S. Patent Application Publication No. 2010/0041054.
  • Human iPSCs also express stem cell markers and are capable of generating cells characteristic of all three germ layers.
  • a “parthenogenetic stem cell” refers to a stem cell arising from parthenogenetic activation of an egg. Methods of creating a parthenogenetic stem cell are known in the art. See, for example, Cibelli et al. (2002) Science 295(5556):819 and Vrana et al. (2003) Proc. Natl. Acad. Sci. USA 100(Suppl. 1)11911-6.
  • pluripotent gene or marker intends an expressed gene or protein that has been correlated with an immature or undifferentiated phenotype, e.g., Oct 3/4, Sox2, Nanog, c-Myc and LIN-28. Methods to identify such are known in the art and systems to identify such are commercially available from, for example, EMD Millipore (MILLIPLEX® Map Kit).
  • hematopoietic stem cells are cells, such as stem cells, that give rise to all types of blood cells, including but not limited to white blood cells, red blood cells, and platelets. Hematopoietic stem cells can be found in the peripheral blood and the bone marrow.
  • an immune cell as disclosed herein is derived from an HSC.
  • phenotype refers to a description of an individual’s trait or characteristic that is measurable and that is expressed only in a subset of individuals within a population.
  • an individual’s phenotype includes the phenotype of a single cell, a substantially homogeneous population of cells, a population of differentiated cells, or a tissue comprised of a population of cells.
  • a population of cells as described herein is substantially homogeneous.
  • substantially homogeneous describes a population of cells in which more than about 50%, or alternatively more than about 60 %, or alternatively more than 70 %, or alternatively more than 75 %, or alternatively more than 80%, or alternatively more than 85 %, or alternatively more than 90%, or alternatively more than 95 %, of the cells are of the same or similar phenotype. Phenotype can be determined by a pre-selected cell surface marker or other marker.
  • isolated refers to molecules, biologicals, cellular materials, cells or biological samples being substantially free from other materials.
  • isolated refers to nucleic acid, such as DNA or RNA, or protein or polypeptide (e.g., an antibody or derivative thereof), or cell or cellular organelle, or tissue or organ, separated from other DNAs or RNAs, or proteins or polypeptides, or cells or cellular organelles, or tissues or organs, respectively, that are present in the natural source.
  • isolated also refers to a nucleic acid or peptide that is substantially free of cellular material, viral material, or culture medium when produced by recombinant DNA techniques, or chemical precursors or other chemicals when chemically synthesized.
  • an “isolated nucleic acid” is meant to include nucleic acid fragments which are not naturally occurring as fragments and would not be found in the natural state.
  • isolated is also used herein to refer to polypeptides which are isolated from other cellular proteins and is meant to encompass both purified and recombinant polypeptides.
  • isolated is also used herein to refer to cells or tissues that are isolated from other cells or tissues and is meant to encompass both cultured and engineered cells or tissues.
  • isolated cell generally refers to a cell that is substantially separated from other cells of a tissue.
  • a population of cells intends a collection of more than one cell that is identical (clonal) or non-identical in phenotype or genotype or both.
  • the population can be purified, highly purified, substantially homogenous or heterogeneous as described herein.
  • “Substantially homogeneous” describes a population of cells in which more than about 50%, or alternatively more than about 60%, or alternatively more than 70%, or alternatively more than 75%, or alternatively more than 80%, or alternatively more than 85%, or alternatively more than 90%, or alternatively, more than 95%, of the cells are of the same or similar phenotype.
  • Phenotype can be determined by a pre-selected cell surface marker or other marker.
  • a purified nucleic acid, peptide, protein, biological complexes, cell or other active compound is one that is isolated in whole or in part from proteins or other contaminants.
  • substantially purified peptides, proteins, biological complexes, cell or other active compounds for use within the disclosure comprise more than 80% of all macromolecular species present in a preparation prior to admixture or formulation of the peptide, protein, biological complex, cell or other active compound with a pharmaceutical carrier, excipient, buffer, absorption enhancing agent, stabilizer, preservative, adjuvant or other co-ingredient in a complete pharmaceutical formulation for therapeutic administration.
  • the peptide, protein, biological complex or other active compound is purified to represent greater than 90%, often greater than 95% of all macromolecular species present in a purified preparation prior to admixture with other formulation ingredients.
  • the purified preparation may be essentially homogeneous, wherein other macromolecular species are not detectable by conventional techniques.
  • the term “engineered” or “recombinant” refers to having at least one modification not normally found in a naturally occurring protein, polypeptide, polynucleotide, strain, wild-type strain or the parental host strain of the referenced species. In some embodiments, the term “engineered” or “recombinant” refers to being synthetized by human intervention.
  • autologous in reference to cells refers to cells that are isolated and infused back into the same subject (recipient or host). “Allogeneic” refers to non-autologous cells.
  • a “composition” as used herein refers to an active agent, such as a compound as disclosed herein and a carrier, inert or active.
  • the carrier can be, without limitation, solid such as a bead or resin, or liquid, such as phosphate buffered saline.
  • a “pharmaceutical composition” is intended to include the combination of an active polypeptide, polynucleotide, antibody or a cell with a carrier, inert or active such as a solid support, making the composition suitable for diagnostic or therapeutic use in vitro, in vivo or ex vivo.
  • the term “pharmaceutically acceptable carrier” encompasses any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, and emulsions, such as an oil/water or water/oil emulsion, and various types of wetting agents.
  • the compositions also can include stabilizers and preservatives.
  • stabilizers and adjuvants see Martin (1975) Remington’s Pharm. Sci., 15th Ed. (Mack Publ. Co., Easton).
  • Administration or treatment in “combination” refers to administering two agents such that their pharmacological effects are manifest at the same time. Combination does not require administration at the same time or substantially the same time, although combination can include such administrations. Such additional agents are described herein, e.g., cytoreductive therapy.
  • Cytoreductive therapy includes but is not limited to chemotherapy, cryotherapy, and radiation therapy. Agents that act to reduce cellular proliferation are known in the art and widely used. Chemotherapy drugs that kill cancer cells only when they are dividing are termed cell-cycle specific. These drugs include agents that act in S-phase, including topoisomerase inhibitors and anti-metabolites.
  • Topoisomerase inhibitors are drugs that interfere with the action of topoisomerase enzymes (topoisomerase I and II). During the process of chemo treatments, topoisomerase enzymes control the manipulation of the structure of DNA necessary for replication, and are thus cell cycle specific. Examples of topoisomerase I inhibitors include the camptothecan analogs listed above, irinotecan and topotecan. Examples of topoisomerase II inhibitors include amsacrine, etoposide, etoposide phosphate, and teniposide.
  • Antimetabolites are usually analogs of normal metabolic substrates, often interfering with processes involved in chromosomal replication. They attack cells at very specific phases in the cycle. Antimetabolites include folic acid antagonists, e.g., methotrexate; pyrimidine antagonist, e.g., 5-fluorouracil, foxuridine, cytarabine, capecitabine, and gemcitabine; purine antagonist, e.g., 6-mercaptopurine and 6-thioguanine; adenosine deaminase inhibitor, e.g., cladribine, fludarabine, nelarabine and pentostatin; and the like.
  • folic acid antagonists e.g., methotrexate
  • pyrimidine antagonist e.g., 5-fluorouracil, foxuridine, cytarabine, capecitabine, and gemcitabine
  • purine antagonist e.g., 6-mercaptopurine and 6-thi
  • Plant alkaloids are derived from certain types of plants.
  • the vinca alkaloids are made from the periwinkle plant (Catharanthus rosea).
  • the taxanes are made from the bark of the Pacific Yew tree (taxus).
  • the vinca alkaloids and taxanes are also known as antimicrotubule agents.
  • the podophyllotoxins are derived from the May apple plant. Camptothecan analogs are derived from the Asian “Happy Tree” (Camptotheca acuminata). Podophyllotoxins and camptothecan analogs are also classified as topoisomerase inhibitors.
  • the plant alkaloids are generally cell-cycle specific.
  • Examples of these agents include vinca alkaloids, e.g., vincristine, vinblastine and vinorelbine; taxanes, e.g., paclitaxel and docetaxel; podophyllotoxins, e.g., etoposide and tenisopide; and camptothecan analogs, e.g., irinotecan and topotecan.
  • vinca alkaloids e.g., vincristine, vinblastine and vinorelbine
  • taxanes e.g., paclitaxel and docetaxel
  • podophyllotoxins e.g., etoposide and tenisopide
  • camptothecan analogs e.g., irinotecan and topotecan.
  • Cryotherapy includes, but is not limited to, therapies involving decreasing the temperature, for example, hypothermic therapy.
  • Radiation therapy includes, but is not limited to, exposure to radiation, e.g., ionizing radiation, UV radiation, as known in the art.
  • exemplary dosages include, but are not limited to, a dose of ionizing radiation at a range from at least about 2 Gy to not more than about 10 Gy and/or a dose of ultraviolet radiation at a range from at least about 5 J/m 2 to not more than about 50 J/m 2 , usually about 10 J/m 2 .
  • a combined therapy may be a drug increasing expression of FLT3 on a cancer cell.
  • the drug comprises an FLT3 inhibitor.
  • FLT3 inhibitor refers to a molecule that binds FLT3 and decreases its activity. Not to be bound by theory, it is believed that such FLT3 inhibitors can increase surface FLT3 expression on cells.
  • FLT3 inhibitors include gilteritinib (Astellas, CID 49803313), quizaritinib (Ambit Biosciences, CID 24889392), midostaurin (Novartis, CID 9829523), sorafenib (Bayer and Onxy Pharmaceuticals, CID 216239), sunitinib (Pfizer, CID 5329102), lestarutinib (Cephalon, CID 126565), FF-10101 (Fuijfilm), dovitinib (Novartis or Oncology Venture, CID 9886808), and equivalents thereof such as but not limited to salts and hydrates, for example, Gilteritinib Fumarate (CID 76970819), Quizartinib Dihydrochloride (CID 25184035), Midostaurin Hydrate (CID 71311854), Sorafenib tosylate (CID 406563), Sora
  • an “effective amount” is an amount sufficient to effect beneficial or desired results.
  • An effective amount can be administered in one or more administrations, applications or dosages. Such delivery is dependent on a number of variables including the time period for which the individual dosage unit is to be used, the bioavailability of the therapeutic agent, the route of administration, etc. It is understood, however, that specific dose levels of the therapeutic agents disclosed herein for any particular subject depends upon a variety of factors including the activity of the specific compound employed, bioavailability of the compound, the route of administration, the age of the animal and its body weight, general health, sex, the diet of the animal, the time of administration, the rate of excretion, the drug combination, and the severity of the particular disorder being treated and form of administration.
  • “Therapeutically effective amount” of a drug or an agent refers to an amount of the drug or the agent that is an amount sufficient to obtain a pharmacological response; or alternatively, is an amount of the drug or agent that, when administered to a patient with a specified disorder or disease, is sufficient to have the intended effect, e.g., treatment, alleviation, amelioration, palliation or elimination of one or more manifestations of the specified disorder or disease in the patient.
  • a therapeutic effect does not necessarily occur by administration of one dose, and may occur only after administration of a series of doses.
  • a therapeutically effective amount may be administered in one or more administrations.
  • contacting means direct or indirect binding or interaction between two or more molecules or other entities.
  • a particular example of direct interaction is binding.
  • a particular example of an indirect interaction is where one entity acts upon an intermediary molecule, which in turn acts upon the second referenced entity.
  • Contacting as used herein includes in solution, in solid phase, in vitro, ex vivo, in a cell and in vivo. Contacting in vivo can be referred to as administering, or administration.
  • administering or “delivery” of a cell or vector or other agent and compositions containing same can be performed in one dose, continuously or intermittently throughout the course of treatment. Methods of determining the most effective means and dosage of administration are known to those of skill in the art and will vary with the composition used for therapy, the purpose of the therapy, the target cell being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician or in the case of animals, by the treating veterinarian. Suitable dosage formulations and methods of administering the agents are known in the art.
  • Route of administration can also be determined and method of determining the most effective route of administration are known to those of skill in the art and will vary with the composition used for treatment, the purpose of the treatment, the health condition or disease stage of the subject being treated, and target cell or tissue.
  • route of administration include intravenous, intracranial, oral administration, intraperitoneal, infusion, nasal administration, inhalation, injection, and topical application.
  • the administration is an intratumoral administration, or administration to a tumor microenvironment, or both.
  • the administration is an infusion (for example to peripheral blood of a subject) over a certain period of time, such as about 30 minutes, about 1 hour, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 7 hours, about 8 hours, about 9 hours, about 10 hours, about 11 hours, about 12 hours, about 24 hours or longer.
  • administration shall include without limitation, administration by oral, parenteral (e.g., intramuscular, intraperitoneal, intravenous, intracerebroventricular (ICV), intrathecal, intracistemal injection or infusion, subcutaneous injection, or implant), by inhalation spray nasal, vaginal, rectal, sublingual, urethral (e.g., urethral suppository) or topical routes of administration (e.g., gel, ointment, cream, aerosol, etc.) and can be formulated, alone or together, in suitable dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants, excipients, and vehicles appropriate for each route of administration.
  • the disclosure is not limited by the route of administration, the formulation or dosing schedule.
  • administering can be performed in one dose, continuously or intermittently throughout the course of treatment. Methods of determining the most effective means and dosage of administration are known to those of skill in the art and will vary with the composition used for therapy, the purpose of the therapy, the target cell being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician. Suitable dosage formulations and methods of administering the agents are known in the art. Route of administration can also be determined and method of determining the most effective route of administration are known to those of skill in the art and will vary with the composition used for treatment, the purpose of the treatment, the health condition or disease stage of the subject being treated, and target cell or tissue.
  • lxlO 4 to lxlO 15 or ranges in between of cells as disclosed herein are administrated to a subject, such as lxlO 7 to lxlO 10 .
  • administering or a grammatical variation thereof also refers to more than one doses with certain interval.
  • the interval is 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 1 week, 10 days, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 1 year or longer.
  • one dose is repeated for once, twice, three times, four times, five times, six times, seven times, eight times, nine times, ten times or more.
  • cells as disclosed herein may be administered to a subject weekly and for up to four weeks.
  • the compositions and therapies can be combined with other therapies, e.g., lymphodepletion chemotherapy followed by infusions (e.g., four weekly infusions) of the therapy, defining one cycle, followed by additional cycles until a partial or complete response is seen or alternatively utilized as a “bridging” therapy to another modality, such as hematopoietic stem cell transplantation or CAR T cell therapy.
  • therapies e.g., lymphodepletion chemotherapy followed by infusions (e.g., four weekly infusions) of the therapy, defining one cycle, followed by additional cycles until a partial or complete response is seen or alternatively utilized as a “bridging” therapy to another modality, such as hematopoietic stem cell transplantation or CAR T cell therapy.
  • An agent of the present disclosure can be administered for therapy by any suitable route of administration. It will also be appreciated that the optimal route will vary with the condition and age of the recipient, and the disease being treated.
  • treating or “treatment” of a disease in a subject refers to (1) preventing the symptoms or disease from occurring in a subject that is predisposed or does not yet display symptoms of the disease; (2) inhibiting the disease or arresting its development; or (3) ameliorating or causing regression of the disease or the symptoms of the disease.
  • treatment is an approach for obtaining beneficial or desired results, including clinical results.
  • beneficial or desired results can include one or more, but are not limited to, alleviation or amelioration of one or more symptoms, diminishment of extent of a condition (including a disease), stabilized (i.e., not worsening) state of a condition (including disease), delay or slowing of condition (including disease), progression, amelioration or palliation of the condition (including disease), states and remission (whether partial or total), whether detectable or undetectable.
  • the disease is cancer
  • the following clinical end points are non-limiting examples of treatment: reduction in tumor burden, slowing of tumor growth, longer overall survival, longer time to tumor progression, inhibition of metastasis or a reduction in metastasis of the tumor.
  • treatment excludes prophylaxis.
  • the term “disease” or “disorder” as used herein refers to a cancer or tumor (which are used interchangeably), a status of being diagnosed with such disease, a status of being suspect of having such disease, or a status of at high risk of having such disease.
  • the disease can be a primary, recurrent, recalcitrant or metastatic cancer.
  • the disease is a cancer.
  • the cancer is a leukemia or a lymphoma.
  • a “leukemia” is a cancer of the blood or bone marrow characterized by an abnormal increase of immature white blood cells.
  • the disease is having a cancer cell expressing FLT3.
  • the cancer is acute myeloid leukemia (AML) or acute lymphoblastic leukemia (ALL).
  • AML acute myeloid leukemia
  • ALL acute lymphoblastic leukemia
  • ALL acute lymphocytic leukemia or acute lymphoid leukemia - is a cancer of the white blood cells, characterized by the overproduction and accumulation of malignant, immature leukocytes (lymphoblasts) resulting a lack of normal, healthy blood cells.
  • a “lymphoma” is a cancer of the blood characterized by the development of blood cell tumors and symptoms of enlarged lymph nodes, fever, drenching sweats, unintended weight loss, itching, and constantly feeling tired.
  • the disease is a solid tumor cancer.
  • Non-limiting examples include sarcomas and carcinomas of the tissues, e.g., brain cancer, renal cancer, breast cancer, adenocarcinoma, neurological cancer, lung cancer, colorectal cancer or glioblastoma (GBM).
  • the disease is selected from breast cancer, melanoma, carcinoid, ovarian cancer, cervical cancer, pancreatic cancer, colorectal cancer, prostate cancer, endometrial cancer, renal caner, glioma, neurological cancer, skin cancer, head and neck cancer, stomach cancer, liver cancer, testis cancer, lung cancer, thyroid cancer, lymphoma, urothelial cancer, or any other cancer as identified in www.proteinatlas.org/ENSG00000122025-FLT3/pathology or Sung Hee Lim et al. Oncotarget. 2017 Jan 10; 8(2): 3237-3245.
  • the disease is myelodysplastic syndromes.
  • cancer as recited in the methods and other disclosure herein may be replaced by the term disease or myelodysplastic syndromes.
  • treatment refers to a patient or individual who has been diagnosed with or is predisposed to a disease as disclosed herein. This patient has not yet developed characteristic disease pathology.
  • a “cancer” is a disease state characterized by the presence in a subject of cells demonstrating abnormal uncontrolled replication and in some aspects, the term may be used interchangeably with the term “tumor.”
  • cancer or tumor antigen refers to an antigen known to be associated and expressed on the surface with a cancer cell or tumor cell or tissue
  • cancer or tumor targeting antibody refers to an antibody that targets such an antigen.
  • first line or “second line” or “third line” refers to the order of treatment received by a patient.
  • First line therapy regimens are treatments given first, whereas second or third line therapy are given after the first line therapy or after the second line therapy, respectively.
  • the National Cancer Institute defines first line therapy as “the first treatment for a disease or condition. In patients with cancer, primary treatment can be surgery, chemotherapy, radiation therapy, or a combination of these therapies. First line therapy is also referred to those skilled in the art as “primary therapy and primary treatment.” See National Cancer Institute website at www.cancer.gov, last visited on May 1, 2008.
  • a patient is given a subsequent chemotherapy regimen because the patient did not show a positive clinical or sub-clinical response to the first line therapy or the first line therapy has stopped.
  • the methods and treatments as described herein can be provides as a first line, second line, third line, fourth line or fifth line therapy.
  • the improved sequence which is also referred to herein as an optimized sequence, provides higher expression level of the CAR as well as the truncated protein marker on a cell, and thus is especially suitable for use in transducing a cell and identifying or isolating the transduced cell. See, e.g., FIGURE 4C v.s. FIGURE 4D.
  • the CAR expressing cells transduced with the optimized sequence demonstrates higher cytotoxicity against tumor cells. See, e.g., FIGURE 5 A.
  • nucleic acid molecule encoding an anti- FMS-like tyrosine kinase 3 (FLT3) chimeric antigen receptor (CAR).
  • the nucleic acid molecule comprises, or alternatively consists essentially of, or yet further consists of the polynucleotide of nucleotide (nt) 55 to nt 777, or nt 1 to nt 777, or the full length of SEQ ID NO: 1 or an equivalent thereof.
  • nucleic acid molecule encoding an anti-FMS-like tyrosine kinase 3 (FLT3) antigen binding fragment comprising, or consisting essentially of, or yet further consisting of the polynucleotide of nucleotide (nt) 55 to nt 777 of SEQ ID NO: 1 or an equivalent there of or the polynucleotide of nt 55 to nt 777 of SEQ ID NO: 12.
  • FLT3 anti-FMS-like tyrosine kinase 3
  • the nucleic acid molecule comprises, or consists essentially of, or yet further consists of the polynucleotide of nt 1 to nt 777 of SEQ ID NO: 1 or an equivalent thereof which is at least 85% identical to nt 1 to nt 777 of SEQ ID NO: 1 or comprising the polynucleotide of nt 1 to nt 777 of SEQ ID NO: 12.
  • the equivalent is at least about 75% (including but not limited to at least about 76%, at least about 77%, at least about 78%, at least about 79%, at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to nucleotide (nt) 55 to nt 777, or nt 1 to nt 777, or the full length of SEQ ID NO: 1 with the proviso that in one aspect, the at least one of optimized nucleotides of SEQ ID NO: 1 are retained in the equivalent. Additionally or alternatively, the equivalent comprises the polyntide (nt) 55 to
  • the nucleic acid molecule or the equivalent thereof comprises, or alternatively consists essentially of, or yet further consists of: a polynucleotide encoding a light chain complementarity-determining region 1 (CDRL1) as set forth in RASQSISNNLH (SEQ ID NO: 15), a polynucleotide encoding a light chain complementarity-determining region 2 (CDRL2) as set forth in YASQSIS (SEQ ID NO: 16), a polynucleotide encoding a light chain complementarity-determining region 3 (CDRL3) as set forth in QQSNTWPYT (SEQ ID NO: 17), a polynucleotide encoding a heavy chain complementarity-determining region 1 (CDRH1) as set forth in SYWMH (SEQ ID NO: 18), a polynucleotide encoding a heavy chain complementarity-determining region 1 (CDRH1) as set forth in SYWMH (SEQ ID NO: 18),
  • the nucleic acid molecule or the equivalent thereof comprises a polynucleotide encoding an antibody heavy chain variable region of QVQLQQPGAELVKPGASLKLSCKSSGYTFTSYWMHWVRQRPGHGLEWIGEIDPSDS YKDYNQKFKDKATLTVDRSSNTAYMHLSSLTSDDSAVYYCARAITTTPFDFWGQGT TLTVSS (SEQ ID NO: 21) and an antibody light chain variable region of DIVLTQSPATLSVTPGDSVSLSCRASQSISNNLHWYQQKSHESPRLLIKYASQSISGIPS RF SGSGSGTDFTLSIN S VETEDF GVYFCQQSNTWPYTF GGGTKLEIKR (SEQ ID NO: 22).
  • the nucleic acid molecule or the equivalent encodes a single chain Fv (scFv) specifically recognizing and binding FLT3, a signal sequence, a spacer, a transmembrane domain, a co-stimulatory domain, and a intracellular signaling domain.
  • scFv single chain Fv
  • the nucleic acid molecule or the equivalent comprises, or consists essentially of, or yet further consists of a polypeptide that encodes a signal peptide, a polypeptide that encodes an FLT3 antigen binding fragment, a polypeptide that encodes a hinge domain, a polypeptide that encodes a transmembrane domain, a polypeptide that encodes a costimulatory signaling region, a polypeptide that encodes an intracellular signaling domain.
  • the nucleic acid molecule or the equivalent comprises, or consists essentially of, or yet further consists of a polypeptide that encodes a signal peptide, a polypeptide that encodes an FLT3 antigen binding fragment, a polypeptide that encodes a hinge domain, a polypeptide that encodes a CD28 transmembrane domain, a polypeptide that encodes a CD28 costimulatory signaling region, a polypeptide that encodes an intracellular signaling domain.
  • the nucleic acid molecule or the equivalent as disclosed herein encodes the polypeptide of amino acid (aa) 19 to aa 259 of SEQ ID NO: 2. In further embodiments, the nucleic acid molecule or the equivalent as disclosed herein encodes the polypeptide of aa 1 to aa 259 of SEQ ID NO: 2. In some embodiments, the nucleic acid molecule or the equivalent encodes the polypeptide of SEQ ID NO: 2.
  • the nucleic acid molecule further comprises a polynucleotide encoding a truncated CD 19, or a truncated EGFR.
  • the truncated EGFR comprises, or alternatively consists essentially of, or yet further consists of the polypeptide of SEQ ID NO: 10.
  • the truncated CD19 comprises, or alternatively consists essentially of, or yet further consists of the polypeptide of SEQ ID NO: 13 or 11.
  • the nucleic acid molecule as disclosed herein further comprises a polynucleotide as set forth in SEQ ID NO: 28 or an equivalent thereof encoding a truncated CD 19.
  • the nucleic acid molecule further comprises a polynucleotide encoding a ribosomal skip sequence located between the polynucleotide encoding the CAR and the polynucleotide encoding the truncated CD 19 or the truncated EGFR. In some embodiments, the nucleic acid molecule further comprises a T2A skip sequence and a sequence encoding the truncated EGFR or the truncated CD 19. In some embodiments, the nucleic acid molecule further comprises a T2A skip sequence and a sequence encoding a truncated EGFR.
  • the nucleic acid molecule further comprises a T2A skip sequence and a sequence encoding a truncated CD 19. In some embodiments, the nucleic acid molecule further comprises a T2A skip sequence and a sequence encoding a truncated EGFR.
  • the nucleic acid molecule as disclosed herein comprises, or alternatively consists essentially of, or yet further consists of an equivalent of SEQ ID NO: 1.
  • the equivalent of SEQ ID NO: 1 consists of about 55% to about 60% of G and C residues, or any percentage or range there between.
  • the equivalent of SEQ ID NO: 1 consists of about 58% of G and C residues.
  • at least about 70% of the codon(s) encoding an alanine (Ala) amino acid residue in the equivalent of SEQ ID NO: 1 consists of GCC.
  • At least about 60% of the codon(s) encoding a cysteine (Cys) amino acid residue in the equivalent of SEQ ID NO: 1 consists of TGC. In some embodiments, at least about 60% of the codon(s) encoding an aspartic acid (Asp) amino acid residue in the equivalent of SEQ ID NO: 1 consists of GAC. In some embodiments, at least about 75% of the codon(s) encoding a glutamic acid (Glu) amino acid residue in the equivalent of SEQ ID NO: 1 consists of GAG.
  • At least about 60% of the codon(s) encoding a phenylalanine (Phe) amino acid residue in the equivalent of SEQ ID NO: 1 consists of TTC. In some embodiments, at least about 55% of the codon(s) encoding a glycine (Gly) amino acid residue in the equivalent of SEQ ID NO: 1 consists of GGC. In some embodiments, at least about 25% of the codon(s) encoding a glycine (Gly) amino acid residue in the equivalent of SEQ ID NO: 1 consists of GGA.
  • At least about 80% of the codon(s) encoding a histidine (His) amino acid residue in the equivalent of SEQ ID NO: 1 consists of CAC. In some embodiments, at least about 65% of the codon(s) encoding an isoleucine (lie) amino acid residue in the equivalent of SEQ ID NO: 1 consists of ATC. In some embodiments, at least about 70% of the codon(s) encoding a lysine (Lys) amino acid residue in the equivalent of SEQ ID NO: 1 consists of AAG.
  • At least about 65% of the codon(s) encoding a leucine (Leu) amino acid residue in the equivalent of SEQ ID NO: 1 consists of CTG. In some embodiments, at least about 90% of the codon(s) encoding a methionine (Met) amino acid residue in the equivalent of SEQ ID NO: 1 consists of ATG. In some embodiments, at least about 55% of the codon(s) encoding an asparagine (Asn) amino acid residue in the equivalent of SEQ ID NO: 1 consists of AAC. In some embodiments, at least about 40% of the codon(s) encoding a proline (Pro) amino acid residue in the equivalent of SEQ ID NO: 1 consists of CCC.
  • At least about 45% of the codon(s) encoding a threonine (Thr) amino acid residue in the equivalent of SEQ ID NO: 1 consists of ACC. In some embodiments, at least about 40% of the codon(s) encoding a Thr amino acid residue in the equivalent of SEQ ID NO: 1 consists of ACA. In some embodiments, at least about 65% of the codon(s) encoding a valine (Val) amino acid residue in the equivalent of SEQ ID NO: 1 consists of GTG. In some embodiments, at least about 90% of the codon(s) encoding a tryptophan (Trp) amino acid residue in the equivalent of SEQ ID NO: 1 consists of TGG.
  • At least about 50% of the codon(s) encoding a tyrosine (Tyr) amino acid residue in the equivalent of SEQ ID NO: 1 consists of TAC. In some embodiments, at least about 45% of the codon(s) encoding a Tyr amino acid residue in the equivalent of SEQ ID NO: 1 consists of TAT.
  • the equivalent of SEQ ID NO: 1 comprises, or alternatively consists essentially of, or yet further consists of the codon consisting of GCG at a frequency of about 0% to about 1% optionally about 0.435%, the codon consisting of GCA at a frequency of about 0% to about 1% optionally about 0.435%, the codon consisting of GCT at a frequency of about 0% to about 1% optionally about 0.652%, the codon consisting of GCC at a frequency of about 3% or higher optionally about 3.913%; the codon consisting of TGT at a frequency of about 0% to about 1% optionally about 0.652%, the codon consisting of TGC at a frequency of about 1% or higher optionally about 1.087%; the codon consisting of GAT at a frequency of about 0% to about 2% optionally about 1.739%, the codon consisting of GAC at a frequency of about 3% or higher optionally about 3.478%; the codon consisting of GAG at
  • the equivalent of SEQ ID NO: 1 comprises a codon frequency or codon usage preference as disclosed in the Table below.
  • the nucleic acid molecule as disclosed herein comprises, or alternatively consists essentially of, or yet further consists of an equivalent of nt 1 to nt 777 of SEQ ID NO: 1.
  • the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of about 55% to about 60% of G and C residues, or any percentage or range there between.
  • the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of about 58% of G and C residues.
  • At least about 80% of the codon(s) encoding an alanine (Ala) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of GCC. In some embodiments, at least about 50% of the codon(s) encoding a cysteine (Cys) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of TGC. In some embodiments, at least about 50% of the codon(s) encoding a cysteine (Cys) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of TGT.
  • At least about 60% of the codon(s) encoding an aspartic acid (Asp) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of GAC.
  • at least about 70% of the codon(s) encoding a glutamic acid (Glu) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of GAG.
  • at least about 55% of the codon(s) encoding a phenylalanine (Phe) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of TTC.
  • At least about 60% of the codon(s) encoding a glycine (Gly) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of GGC. In some embodiments, at least about 30% of the codon(s) encoding a glycine (Gly) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of GGA. In some embodiments, at least about 80% of the codon(s) encoding a histidine (His) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of CAC.
  • At least about 80% of the codon(s) encoding an isoleucine (lie) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of ATC. In some embodiments, at least about 80% of the codon(s) encoding a lysine (Lys) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of AAG. In some embodiments, at least about 90% of the codon(s) encoding a leucine (Leu) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of CTG.
  • At least about 90% of the codon(s) encoding a methionine (Met) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of ATG.
  • at least about 50% of the codon(s) encoding an asparagine (Asn) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of AAC.
  • at least about 50% of the codon(s) encoding an asparagine (Asn) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of AAT.
  • At least about 40% of the codon(s) encoding a proline (Pro) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of CCC.
  • at least about 90% of the codon(s) encoding a glutamine (Gin) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of CAG.
  • at least about 50% of the codon(s) encoding an arginine (Arg) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of CGG.
  • At least about 20% of the codon(s) encoding an Arg amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of AGG.
  • at least about 60% of the codon(s) encoding a serine (Ser) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of AGC.
  • at least about 45% of the codon(s) encoding a threonine (Thr) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of ACC.
  • At least about 45% of the codon(s) encoding a Thr amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of ACA. In some embodiments, at least about 70% of the codon(s) encoding a valine (Val) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO:
  • At least about 90% of the codon(s) encoding a tryptophan (Trp) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of TGG.
  • at least about 60% of the codon(s) encoding a tyrosine (Tyr) amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of TAT.
  • at least about 30% of the codon(s) encoding a Tyr amino acid residue in the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 consists of TAT.
  • the equivalent of nt 1 to nt 777 of SEQ ID NO: 1 comprises, or alternatively consists essentially of, or yet further consists of the codon consisting of GCG at a frequency of about 0% to about 1% optionally about 0.000%, the codon consisting of GCA at a frequency of about 0% to about 1% optionally about 0.000%, the codon consisting of GCT at a frequency of about 0% to about 1% optionally about 0.772%, the codon consisting of GCC at a frequency of about 3% or higher optionally about 3.861%; the codon consisting of TGT or TGC at a frequency of about 0% to about 1% optionally about 0.772%, the codon consisting of TGT or TGC at a frequency of about 0.5% or higher optionally about 0.772%; the codon consisting of GAT at a frequency of about 0% to about 2% optionally about 1.544%, the codon consisting of GAC at a frequency of about 3% or higher
  • nt 1 to nt 777 of SEQ ID NO: 1 comprises a codon frequency or codon usage preference as disclosed in the Table below.
  • the nucleic acid molecule as disclosed herein comprises, or alternatively consists essentially of, or yet further consists of an equivalent of nt 55 to nt 777 of SEQ ID NO: 1.
  • the equivalent of SEQ ID NO: 1 comprises nucleotide(nt) 55 to nt 408 of SEQ ID NO: 1. In some embodiments, the equivalent of SEQ ID NO: 1 comprises nt 454 to nt 777 of SEQ ID NO: 1. In some embodiments, the equivalent of SEQ ID NO: 1 comprises nt 841 to nt 921 of SEQ ID NO: 1. In some embodiments, the equivalent of SEQ ID NO: 1 comprises nt 922 to nt 1044 of SEQ ID NO: 1. In some embodiments, the equivalent of SEQ ID NO: 1 comprises nt 1045 to nt 1380 of SEQ ID NO: 1.
  • the equivalent of SEQ ID NO: 1 comprises nt 55 to nt 408 of SEQ ID NO: 1 and nt 454 to nt 777 of SEQ ID NO: 1. In some embodiments, the equivalent of SEQ ID NO: 1 comprises nt 55 to nt 408 of SEQ ID NO: 1, nt 454 to nt 777 of SEQ ID NO:
  • the nucleic acid molecule comprises, or alternatively consists essentially of, or yet further consists of SEQ ID NO: 1.
  • the equivalent of SEQ ID NO: 1 comprises, or alternatively consists essentially of, or yet further consists of the polynucleotide of SEQ ID NO: 14 comprising at least one nt residue substituted with the aligned nt residue of SEQ ID NO: 1.
  • the substituted nt residue is different from the original nt residue.
  • nucleic acid molecule that encodes the CAR.
  • the nucleic acid molecule further comprise the necessary regulatory sequences, e.g., a promoter for expression in a cell, or an enhancer.
  • the nucleic acid molecule further comprise a first regulatory sequence directing the expression of the CAR.
  • the regulatory sequences comprises one or more of the following: a promoter, an intron, an enhancer, or a polyadenylation signal.
  • the promoter is an EF1 alpha promoter.
  • the promoter is a CMV promoter.
  • the promoter is a MMLV promoter.
  • the nucleic acid molecule further encode a truncated protein marker that may be regulated from the same first regulatory sequence or a second regulatory sequence, such as a promoter element.
  • the second promoter comprises an EF1 alpha promoter.
  • the promoter(s) are selected for the host expression system and will vary with the host and the expression vector and intended use. In one embodiment, any one or both of the regulatory sequences can be cell specific or tissue specific.
  • the CAR described herein can be produced by any means known in the art, though preferably it is produced using recombinant DNA techniques.
  • Nucleic acids encoding the several regions of the chimeric receptor can be prepared and assembled into a complete coding sequence by standard techniques of molecular cloning known in the art (genomic library screening, overlapping PCR, primer-assisted ligation, site-directed mutagenesis, etc.) as is convenient.
  • the resulting coding region is preferably inserted into an expression vector and used to transform a suitable expression host cell line, preferably a T lymphocyte, and most preferably an autologous T lymphocyte.
  • Central memory T cells are one useful T cell subset.
  • Central memory T cell can be isolated from peripheral blood mononuclear cells (PBMC) by selecting for CD45RO+/CD62L+ cells, using, for example, the CLINIMACS® device to immunomagnetically select cells expressing the desired receptors.
  • the cells enriched for central memory T cells can be activated with anti-CD3/CD28, transduced with, for example, a lentiviral vector that directs the expression of the CAR as well as a non-immunogenic surface marker for in vivo detection, ablation, and potential ex vivo selection.
  • the activated/genetically modified CAR T cells can be expanded in vitro with IL-2/IL-15 and then cryopreserved. Additional methods of preparing CAR T cells can be found in PCT/US2016/043392.
  • FLT3 CDR domain amino acid sequences are described in US Patent No. 10,961,312, W02020/010284, Tables 1-4 of the US Patent Application No.: US20180346601, Table V of US Patent Application No.: US20180037657, Table 10 of US Patent Application No.: US20170037149, Table V of US Patent Application No. : US20160272716, Tables 1-3 of US Patent Application No. : US20110091470 and Tables 1-3 of US Patent Application No.: US20090297529.
  • Nonlimiting examples of FLT3 heavy chain variable region and light chain variable region amino acid sequences are described in US Patent No.: 10,961,312, W02020/010284, Tables 1 and 3 of the US Patent Application No.: US20180346601, Table X of US Patent Application No.: US20180037657, Table 10 of US Patent Application No.: US20170037149 and Table VII of US Patent Application No.: US20160272716.
  • the polynucleotide further encodes a transmembrane domain that comprises, or consists essentially of, or yet further consists of, a CD28 transmembrane domain or a CD8 a transmembrane domain.
  • the polynucleotide further encodes an intracellular domain that comprises, or alternatively consists essentially of, or yet further consists of, a CD28 costimulatory signaling region or a 4-1BB costimulatory signaling region or both.
  • the polynucleotide further comprises a polynucleotide that encodes a CD3 zeta signaling domain.
  • a CAR or a cytoplasmic domain thereof as disclosed herein further comprises an IL2R ⁇ or a fragment thereof.
  • the fragments of IL2R ⁇ comprises, or alternatively consists essentially of, or yet consists of a JAK-STAT activation domain of the IL2R ⁇ , facilitating activation of the immune cell.
  • the CAR expressing cell further expresses and optionally secrets an immunoregulatory molecular or a cytokine.
  • the CAR expressing cell comprises a lower expression level or a lower activity of a suppressor of the immunoeregulatory molecular or a cytokine.
  • the CAR expressing cell expresses IL-15 but does not express cytokine-inducible Src homology 2-containing protein (CIS). See, for example, Daher et al. Blood 2021 Feb 4;137(5):624-636.
  • the CAR expressing cell may also comprise a switch mechanism for controlling expression or activation or both expression and activation of the CAR.
  • a CAR may comprise, or consist of, or consist essentially of an extracellular, transmembrane, and intracellular domain, in which the extracellular domain comprises a target-specific binding element that binds a label, binding domain, or tag that is specific for a molecule other than the target antigen that is expressed on or by a target cell (such as a cancer cell).
  • a target cell such as a cancer cell.
  • such label, binding domain or tag recognizes and binds the target antigen that is expressed on or by the target cell.
  • the specificity of the CAR is provided by a second construct that comprises, consists, or consists essentially of a target antigen binding domain and a domain on the CAR that is recognized by or binds to the label, binding domain, or tag.
  • a second construct that comprises, consists, or consists essentially of a target antigen binding domain and a domain on the CAR that is recognized by or binds to the label, binding domain, or tag.
  • a T-cell, NK cell or other cells that express the CAR can be administered to a subject, but it cannot bind a target antigen (i.e., FLT3) until a second composition comprising the label, binding domain, or tag, such as an FLT3-specific binding domain is administered.
  • a target antigen i.e., FLT3
  • CARs of the present disclosure may likewise require multimerization in order to active their function (see, e.g., US 2015/0368342, US 2016/0175359, US 2015/0368360) and/or an exogenous signal, such as a small molecule drug (US 2016/0166613, Yung et al., Science, 2015) in order to elicit an immune cell response, such as a T-cell response or a NK cell response.
  • an exogenous signal such as a small molecule drug (US 2016/0166613, Yung et al., Science, 2015) in order to elicit an immune cell response, such as a T-cell response or a NK cell response.
  • the disclosed CAR expressing cell can comprise a “suicide switch” to induce cell death of the CAR expressing cells following treatment (Buddee et al., PLoS One, 2013) or to downregulate expression of the CAR following binding to the target antigen (WO 2016/011210).
  • a non-limiting exemplary suicide switch or suicide gene is iCasp.
  • a CAR and/or a cytoplasmic domain thereof as disclosed herein further comprise a suicide gene product.
  • the suicide gene product is selected from one or more of: HSV-TK (Herpes simplex virus thymidine kinase), cytosine deaminase, nitroreductase, carboxylesterase, cytochrome P450 or PNP (Purine nucleoside phosphorylase), truncated EGFR, or inducible caspase (“iCasp”).
  • HSV-TK Herpes simplex virus thymidine kinase
  • cytosine deaminase cytosine deaminase
  • nitroreductase carboxylesterase
  • cytochrome P450 or PNP Purine nucleoside phosphorylase
  • truncated EGFR or inducible caspase (“iCasp”).
  • the suicide gene product is a truncated protein maker as disclosed herein.
  • the CD3 ⁇ signaling domain can be followed by a ribosomal skip sequence (e g., LEGGGEGRGSLLTCGD VEENPGPR; SEQ ID NO: 9) and a truncated EGFR having a sequence that is at least 90%, at least 95%, at least 98% identical to or identical to:
  • a ribosomal skip sequence e g., LEGGGEGRGSLLTCGD VEENPGPR; SEQ ID NO: 9
  • a truncated EGFR having a sequence that is at least 90%, at least 95%, at least 98% identical to or identical to:
  • the CD3 ⁇ signaling domain can be followed by a ribosomal skip sequence (e g., LEGGGEGRGSLLTCGD VEENPGPR; SEQ ID NO: 9) and a truncated CD 19 having a sequence that is at least 90%, at least 95%, at least 98% identical to or identical to:
  • a ribosomal skip sequence e g., LEGGGEGRGSLLTCGD VEENPGPR; SEQ ID NO: 9
  • a truncated CD 19 having a sequence that is at least 90%, at least 95%, at least 98% identical to or identical to:
  • This disclosure also provides a vector that comprises, or alternatively consists essentially of, or yet further consists of any one or more of the polynucleotides as disclosed herein.
  • the polynucleotide or vector further comprises, or consists essentially of, or yet further consists of, a regulatory element to drive expression of the polynucleotide or the CAR.
  • the regulatory element comprises one or more of the following: a promoter, an intron, an enhancer, or a polyadenylation signal.
  • the vector further comprises a detectable or purification marker.
  • a vector comprising, or alternatively consisting essentially of, or yet further consisting of a nucleic acid molecule as disclosed herein or a complementary nucleic acid molecule thereof.
  • the vector is a viral vector. Additionally or alternatively, the vector is an expression vector.
  • the vector is a lentiviral vector. In some embodiments, the vector is a retroviral vector.
  • the expression of the nucleotide sequence of SEQ ID NO: 1 or the equivalent thereof is under the control of a promoter, optionally an EF- 1 alpha promoter, a CMV promoter, or a MMLV promoter.
  • the vector is a non-viral vector (such as a plasmid).
  • the vector is a viral vector, non-limiting examples of such are selected from the group of a retroviral vector, a lentiviral vector, an adenoviral vector, and an adeno- associated viral vector.
  • the nucleic acid molecule as disclosed herein can be inserted into a vector, such as an expression vector, e.g., a lentiviral vector or retroviral vector (between the 5’ and 3’ LTRs) or an adenovirus vector or any other vectors that can express a gene from.
  • the vector is derived from or based on a wild-type virus. In further embodiments, the vector is derived from or based on a wild-type lentivirus. Examples of such include without limitation, human immunodeficiency virus (HIV), equine infectious anemia virus (EIAV), visna/maedi virus (VMV), caprine arthritis-encephalitis virus (CAEV), equine infectious anemia virus (EIAV), simian immunodeficiency virus (SIV), bovine immunodeficiency virus (BIV) and feline immunodeficiency virus (FIV).
  • HCV human immunodeficiency virus
  • EIAV equine infectious anemia virus
  • VMV visna/maedi virus
  • CAEV caprine arthritis-encephalitis virus
  • EIAV simian immunodeficiency virus
  • BIV bovine immunodeficiency virus
  • FIV feline immunodeficiency virus
  • retroviral vectors for use in this disclosure include, but are not limited to Invitrogen’s pLenti series versions 4, 6, and 6.2 “ViraPower” system.
  • a viral vector according to the disclosure need not be confined to the components of a particular virus.
  • the viral vector may comprise components derived from two or more different viruses, and may also comprise synthetic components.
  • Vector components can be manipulated to obtain desired characteristics, such as target cell specificity.
  • U.S. Patent No. 6,924,123 discloses that certain retroviral sequence facilitate integration into the target cell genome.
  • each retroviral genome comprises genes called gag, pol and env which code for virion proteins and enzymes. These genes are flanked at both ends by regions called long terminal repeats (LTRs).
  • LTRs are responsible for proviral integration, and transcription. They also serve as enhancer-promoter sequences. In other words, the LTRs can control the expression of the viral genes.
  • Encapsidation of the retroviral RNAs occurs by virtue of a psi sequence located at the 5' end of the viral genome.
  • the LTRs themselves are identical sequences that can be divided into three elements, which are called U3, R and U5.
  • U3 is derived from the sequence unique to the 3' end of the RNA.
  • R is derived from a sequence repeated at both ends of the RNA
  • U5 is derived from the sequence unique to the 5'end of the RNA.
  • the sizes of the three elements can vary considerably among different retroviruses.
  • the site of poly (A) addition (termination) is at the boundary between R and U5 in the right hand side LTR.
  • U3 contains most of the transcriptional control elements of the provirus, which include the promoter and multiple enhancer sequences responsive to cellular and in some cases, viral transcriptional activator proteins.
  • gag encodes the internal structural protein of the virus.
  • Gag protein is proteolytically processed into the mature proteins MA (matrix), CA (capsid) and NC (nucleocapsid).
  • the pol gene encodes the reverse transcriptase (RT), which contains DNA polymerase, associated RNase H and integrase (IN), which mediate replication of the genome.
  • RT reverse transcriptase
  • I integrase
  • the vector RNA genome is expressed from a DNA construct encoding it, in a host cell.
  • the components of the particles not encoded by the vector genome are provided in trans by additional nucleic acid sequences (the "packaging system", which usually includes either or both of the gag/pol and env genes) expressed in the host cell.
  • the set of sequences required for the production of the viral vector particles may be introduced into the host cell by transient transfection, or they may be integrated into the host cell genome, or they may be provided in a mixture of ways. The techniques involved are known to those skilled in the art.
  • a vector for expression of a nucleic acid molecule as disclosed herein in a cell i.e., an expression vector.
  • a vector for producing an expression vector i.e., a production vector or a packaging vector, such as a plasmid for duplicating a viral vector genome, and such vector genome after appropriate packaging into the viral vector is able to transduce a cell resulting in expression of the nucleic acid molecule in the cell.
  • Two exemplified production vectors are illustrated in FIGURE 2 and FIGURE 3.
  • the isolated nucleic acids can be packaged into a retroviral packaging system by using a packaging vector and cell lines.
  • the packaging vector includes, but is not limited to retroviral vector, lentiviral vector, adenoviral vector, and adeno-associated viral vector.
  • the packaging vector contains elements and sequences that facilitate the delivery of genetic materials into cells.
  • the retroviral constructs are packaging vectors comprising at least one retroviral helper DNA sequence derived from a replication- incompetent retroviral genome encoding in trans all virion proteins required to package a replication incompetent retroviral vector, and for producing virion proteins capable of packaging the replication-incompetent retroviral vector at high titer, without the production of replication-competent helper virus.
  • the retroviral DNA sequence lacks the region encoding the native enhancer and/or promoter of the viral 5' LTR of the virus, and lacks both the psi function sequence responsible for packaging helper genome and the 3'
  • the retrovirus is a leukemia virus such as a Moloney Murine Leukemia Virus (MMLV), the Human Immunodeficiency Virus (HIV), or the Gibbon Ape Leukemia virus (GALV).
  • MMLV Moloney Murine Leukemia Virus
  • HSV Human Immunodeficiency Virus
  • GALV Gibbon Ape Leukemia virus
  • the foreign enhancer and promoter may be the human cytomegalovirus (HCMV) immediate early (IE) enhancer and promoter, the enhancer and promoter (U3 region) of the Moloney Murine Sarcoma Virus (MMSV), the U3 region of Rous Sarcoma Virus (RSV), the U3 region of Spleen Focus Forming Virus (SFFV), or the HCMV IE enhancer joined to the native Moloney Murine Leukemia Virus (MMLV) promoter.
  • HCMV human cytomegalovirus
  • IE immediate early
  • IE Enhancr and promoter
  • U3 region of the Moloney Murine Sarcoma Virus
  • RSV Rous Sarcoma Virus
  • SFFV Spleen Focus Forming Virus
  • HCMV IE enhancer joined to the native Moloney Murine Leukemia Virus
  • the retroviral packaging vector may consist of two retroviral helper DNA sequences encoded by plasmid based expression vectors, for example where a first helper sequence contains a cDNA encoding the gag and pol proteins of ecotropic MMLV or GALV and a second helper sequence contains a cDNA encoding the env protein.
  • the Env gene which determines the host range, may be derived from the genes encoding xenotropic, amphotropic, ecotropic, polytropic (mink focus forming) or 10A1 murine leukemia virus env proteins, or the Gibbon Ape Leukemia Virus (GALV env protein, the Human Immunodeficiency Virus env (gpl60) protein, the Vesicular Stomatitus Virus (VSV) G protein, the Human T cell leukemia (HTLV) type I and II env gene products, chimeric envelope gene derived from combinations of one or more of the aforementioned env genes or chimeric envelope genes encoding the cytoplasmic and transmembrane of the aforementioned env gene products and a monoclonal antibody directed against a specific surface molecule on a desired target cell.
  • GLV env protein Gibbon Ape Leukemia Virus
  • gpl60 Human Immunodeficiency Virus env
  • VSV Ves
  • the packaging vectors and retroviral vectors are transiently co-transfected into a first population of mammalian cells that are capable of producing virus, such as human embryonic kidney cells, for example 293 cells (ATCC No. CRL1573, ATCC, Rockville, Md.) to produce high titer recombinant retrovirus-containing supernatants.
  • virus such as human embryonic kidney cells, for example 293 cells (ATCC No. CRL1573, ATCC, Rockville, Md.) to produce high titer recombinant retrovirus-containing supernatants.
  • this transiently transfected first population of cells is then co-cultivated with mammalian target cells, for example human lymphocytes, to transduce the target cells with the foreign gene at high efficiencies.
  • mammalian target cells for example human lymphocytes
  • the supernatants from the above described transiently transfected first population of cells are incubated with mammalian target cells, for example human lymphocytes or hematopoietic stem cells, to transduce the target cells with the foreign gene at high efficiencies.
  • mammalian target cells for example human lymphocytes or hematopoietic stem cells
  • the packaging vectors are stably expressed in a first population of mammalian cells that are capable of producing virus, such as human embryonic kidney cells, for example 293 cells.
  • Retroviral or lentiviral vectors are introduced into cells by either co-transfection with a selectable marker or infection with pseudotyped virus. In both cases, the vectors integrate.
  • vectors can be introduced in an episomally maintained plasmid. High titer recombinant retrovirus-containing supernatants are produced.
  • RNA electroporation As is apparent, when used clinically in a human patient, marker or purification tags will be omitted from the construct.
  • the cells can be transduced using the viral vectors as described herein or alternatively using technology described in Riet et al. (2013) Meth. Mol. Biol. 969: 187-201 entitled “Nonviral RNA transfection to transiently modify T cell with chimeric antigen receptors for adoptive therapy.”
  • Further methods of introducing exogenous nucleic acids into the art are known and include but are not limited to gene delivery using one or more of RNA electroporation, nanotechnology, sleeping beauty vectors, retroviruses, and/or adenoviruses.
  • assays include, for example, "molecular biological” assays well known to those of skill in the art, such as Southern and Northern blotting, RT-PCR and PCR; "biochemical” assays, such as detecting the presence or absence of a particular peptide, e.g., by immunological means (ELISAs and Western blots) or by assays described herein to identify agents falling within the scope of the disclosure.
  • molecular biological assays well known to those of skill in the art, such as Southern and Northern blotting, RT-PCR and PCR
  • biochemical assays, such as detecting the presence or absence of a particular peptide, e.g., by immunological means (ELISAs and Western blots) or by assays described herein to identify agents falling within the scope of the disclosure.
  • a population of human T or NK cells comprising a nucleic acid molecule as disclosed herein or a vector as disclosed herein.
  • the isolated cell or the population of human T orNK cells comprise central memory T cells, NK cells, naive memory T cells, pan T cells, or PBMC substantially depleted for CD25+ cells and CD14+ cells.
  • an isolated cell comprising a nucleic acid molecule as disclosed herein or a vector as disclosed herein.
  • the cell is selected from the group of: an immune cell, an NK cell, a T cell, a stem cell, a progenitor cell or a precursor cell.
  • a population of cells comprising the isolated cell.
  • composition comprising, or consisting essentially of, or yet further consisting of a cell population as disclosed herein a cell as disclosed herein, and a carrier, and optionally a stabilizer, preservative or cryopreservative.
  • the cell population is substantially homogenous.
  • the population comprises at least 90% (including but not limited to at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) of cells of the same at least one cell subtype, e.g., at least 90% of the cells are T cells or NK cells.
  • an isolated cell comprising, or alternatively consisting essentially of, or yet further consisting of, any one or more of: a vector as disclosed herein, a CAR as disclosed herein, a truncated protein maker as disclosed herein, or a polynucleotide as described herein.
  • the cell can be a prokaryotic cell (e.g., a bacterial cell) or a eukaryotic cell.
  • eukaryotic cells include, but are not limited to, a yeast cell, an animal cell, a mammalian cell, a bovine cell, a feline cell, a canine cell, a murine cell, an equine cell, or a human cell.
  • the isolated cell expresses the CAR.
  • the isolated cell expresses the truncated protein marker.
  • the cell further comprise a detectable or purification marker.
  • the eukaryotic cell, mammalian or human cell is an immune cell, optionally a T-cell, a B cell, aNK cell, an NKT cell, a dendritic cell, a myeloid cell, a monocyte, a macrophage, any subsets thereof, or any other immune cell.
  • the cell is an immune cell optionally selected from a T-cell, a B cell, an NK cell, an NKT cell, a dendritic cell, a myeloid cell, a monocyte, a macrophage.
  • the isolated cell is a T-cell, e.g., an animal T-cell, a mammalian T-cell, a feline T-cell, a canine T-cell or a human T-cell.
  • the isolated cell is an NK- cell, e.g., an animal NK-cell, a mammalian NK-cell, a feline NK-cell, a canine NK-cell or a human NK-cell.
  • the isolated cell is an NKT-cell, e.g., an animal NKT-cell, a mammalian NKT-cell, a feline NKT-cell, a canine NKT-cell or a human NKT- cell.
  • the isolated cell is a B-cell, e.g., an animal B-cell, a mammalian B-cell, a feline B-cell, a canine B-cell or a human B-cell. It is appreciated that the same or similar embodiments for each species apply with respect to dendritic cells, myeloid cells, monocytes, macrophages, any subsets of these or the T-cells, NK-cells, NKT- cells, and B-cells as described, or any other immune cells.
  • the T cell is a gamma-delta T cell.
  • the cell is a T cell that has been modified to remove CD52 expression using gene editing technology, e.g., CRISPR or TALEN.
  • the cell is selected from a Hematopoietic stem cell (HSC), an induced pluripotent stem cell (iPSCs), or an immune cell.
  • the immune cell is derived from hematopoietic stem cells (HSCs) or induced pluripotent stem cells (iPSCs).
  • the cells may be derived from patients, donors, or cell lines, such as those available off-the-shelf.
  • the cells can be autologous or allogeneic to the subject being treated.
  • cells prior to expansion and genetic modification of the cells disclosed herein, cells may be obtained from a subject - for instance, in embodiments involving autologous therapy - or a commercially available cell line or culture, or a stem cell such as an induced pluripotent stem cell (iPSC).
  • iPSC induced pluripotent stem cell
  • Cells can be obtained from a number of sources in a subject, including peripheral blood, peripheral blood mononuclear cells, bone marrow, lymph node tissue, cord blood, thymus tissue, tissue from a site of infection, ascites, pleural effusion, spleen tissue, and tumors.
  • Particular subpopulations of immune cells may be isolated through the use of beads or other binding agents available in such kits specific to unique cell surface markers.
  • MACSTM CD4+ and CD8+ MicroBeads may be used to isolate CD4+ and CD8+ T-cells.
  • Alternate non-limiting examples of cells that may be isolated according to known techniques include bulked T-cells, NK T-cells, and gamma delta T-cells.
  • cells may be obtained through commercially available cell cultures, including but not limited to, for T-cells, lines BCL2 (AAA) Jurkat (ATCC® CRL- 2902TM), BCL2 (S70A) Jurkat (ATCC® CRL-2900TM), BCL2 (S87A) Jurkat (ATCC® CRL- 2901TM), BCL2 Jurkat (ATCC® CRL-2899TM), Neo Jurkat (ATCC® CRL-2898TM); for B cells, lines AHH-1 (ATCC® CRL-8146TM), BC-1 (ATCC® CRL-2230TM), BC-2 (ATCC® CRL-2231TM), BC-3 (ATCC® CRL-2277TM), CA46 (ATCC® CRL-1648TM), DG-75 [D.G.- 75] (ATCC® CRL-2625TM), DS-1 (ATCC® CRL-11102TM), EB-3 [EB3] (ATCC® CCL- 85TM), Z-138 (ATCC #CRL-3001),
  • T-cell lines e.g., Deglis, EBT-8, HPB-MLp-W, HUT 78, HUT 102, Karpas 384, Ki 225, My-La, Se-Ax, SKW-3, SMZ-1 and T34; immature T- cell lines, e.g., ALL-SIL, Be13, CCRF-CEM, CML-T1, DND-41, DU.528, EU-9, HD-Mar, HPB-ALL, H-SB2, HT-1, JK-T1, Jurkat, Karpas 45, KE-37, KOPT-K1, K-Tl, L-KAW, Loucy, MAT, MOLT-1, MOLT 3, MOLT-4, MOLT 13, MOLT- 16, MT-1, MT-ALL, P12/Ichikawa, Peer, PER0117, PER-255, PF-382, PFI-285, RPMI-8402, ST-4, SUP-T1 to T14,
  • immature T- cell lines
  • Null leukemia cell lines including but not limited to REH, NALL-1, KM-3, L92-221, are a another commercially available source of immune cells, as are cell lines derived from other leukemias and lymphomas, such as K562 erythroleukemia, THP-1 monocytic leukemia, U937 lymphoma, HEL erythroleukemia, HL60 leukemia, HMC- 1 leukemia, KG-1 leukemia, U266 myeloma.
  • Non-limiting exemplary sources for such commercially available cell lines include the American Type Culture Collection, or ATCC, (atcc.org/) and the German Collection of Microorganisms and Cell Cultures (dsmz.de/).
  • T cells expressing the disclosed CARs may be further modified to reduce or eliminate expression of endogenous TCRs. Reduction or elimination of endogenous TCRs can reduce off-target effects and increase the effectiveness of the T cells.
  • T cells stably lacking expression of a functional TCR may be produced using a variety of approaches. T cells internalize, sort, and degrade the entire T cell receptor as a complex, with a half-life of about 10 hours in resting T cells and 3 hours in stimulated T cells (von Essen, M. et al. 2004. J. Immunol. 173:384-393). Proper functioning of the TCR complex requires the proper stoichiometric ratio of the proteins that compose the TCR complex.
  • TCR function also requires two functioning TCR zeta proteins with IT AM motifs.
  • the activation of the TCR upon engagement of its MHC-peptide ligand requires the engagement of several TCRs on the same T cell, which all must signal properly.
  • TCR expression may eliminated using RNA interference (e.g., shRNA, siRNA, miRNA, etc.), CRISPR, or other methods that target the nucleic acids encoding specific TCRs (e.g., TCR-a and TCR-b) and/or CD3 chains in primary T cells.
  • the T cell By blocking expression of one or more of these proteins, the T cell will no longer produce one or more of the key components of the TCR complex, thereby destabilizing the TCR complex and preventing cell surface expression of a functional TCR. Even though some TCR complexes can be recycled to the cell surface when RNA interference is used, the RNA (e.g., shRNA, siRNA, miRNA, etc.) will prevent new production of TCR proteins resulting in degradation and removal of the entire TCR complex, resulting in the production of a T cell having a stable deficiency in functional TCR expression.
  • shRNA shRNA, siRNA, miRNA, etc.
  • RNAs e.g., shRNA, siRNA, miRNA, etc.
  • expression of inhibitory RNAs in primary T cells can be achieved using any conventional expression system, e.g., a lentiviral expression system.
  • lentiviruses are useful for targeting resting primary T cells, not all T cells will express the shRNAs. Some of these T cells may not express sufficient amounts of the RNAs to allow enough inhibition of TCR expression to alter the functional activity of the T cell.
  • T cells that retain moderate to high TCR expression after viral transduction can be removed, e.g., by cell sorting or separation techniques, so that the remaining T cells are deficient in cell surface TCR or CD3, enabling the expansion of an isolated population of T cells deficient in expression of functional TCR or CD3.
  • CRISPR in primary T cells can be achieved using conventional CRISPR/Cas systems and guide RNAs specific to the target TCRs. Suitable expression systems, e.g. lentiviral or adenoviral expression systems are known in the art. Similar to the delivery of inhibitor RNAs, the CRISPR system can be used to specifically target resting primary T cells or other suitable immune cells for CAR cell therapy. Further, to the extent that CRISPR editing is unsuccessful, cells can be selected for success according to the methods disclosed above.
  • T cells that retain moderate to high TCR expression after viral transduction can be removed, e.g., by cell sorting or separation techniques, so that the remaining T cells are deficient in cell surface TCR or CD3, enabling the expansion of an isolated population of T cells deficient in expression of functional TCR or CD3.
  • a CRISPR editing construct may be useful in both knocking out the endogenous TCR and knocking in the CAR constructs disclosed herein. Accordingly, it is appreciated that a CRISPR system can be designed for to accomplish one or both of these purposes.
  • a cell population comprising, or alternatively consisting essentially of, or yet consisting of a cell as disclosed herein.
  • the cell population is substantially homogenous.
  • the cell population comprises, or alternatively consists essentially of, or yet consists of a Hematopoietic stem cell (HSC), an induced pluripotent stem cell (iPSCs), or an immune cell.
  • the immune cells is selected from a group consisting of T-cells, B cells, NK cells, NKT cells, dendritic cells, myeloid cells, monocytes, or macrophages.
  • the immune cell is derived from HSCs or iPSCs.
  • the cell is a T cell that has been modified to remove CD52 expression using gene editing technology, e.g., CRISPR or TALEN.
  • a method of preparing CAR T or NK cells comprises, or alternatively consists essentially of, or yet further consists of providing a population of autologous or allogeneic human T or NK cells and transducing the T or NK cells by a vector as disclosed herein or a nucleic acid molecule as disclosed herein.
  • a method of preparing a CAR expressing cell comprises, or alternatively consists essentially of, or yet further consists of transducing the cell with a vector as disclosed herein or a nucleic acid molecule as disclosed herein.
  • the disclosure is drawn to a method of producing a CAR- expressing cell comprising transducing an isolated cell with any of the polynucleotides as disclosed herein.
  • the method further comprises selecting and isolating the cell expressing the CAR.
  • the method of producing a CAR expressing cell further comprises, or alternatively consists essentially of, or yet further consists of activating and expanding the population of CAR expressing cells.
  • the cells can be activated and expanded using generally known methods such as those described in U.S. Patent Nos. 6,352,694; 6,534,055; 6,905,680; 6,692,964;
  • soluble ligands and/or cytokines, or stimulating agents may be required to activate and expand cells.
  • the relevant reagents are well known in the art and are selected according to known immunological principles. For instance, soluble CD-40 ligand may be helpful in activating and expanding certain B-cell populations; similarly, irradiated feeder cells may be used in the procedure for activation and expansion of NK cells.
  • Isolation methods for use in relation to this disclosure include, but are not limited to Life Technologies Dynabeads® System activation and expansion kits; BD Biosciences PhosflowTM activation kits, Miltenyi Biotec MACSTM activation/expansion kits, and other commercially available cell kits specific to activation moieties of the relevant cell.
  • Particular subpopulations of immune cells may be activated or expanded through the use of beads or other agents available in such kits.
  • a-CD3/a-CD28 Dynabeads® may be used to activate and expand a population of isolated T-cells.
  • the FLT3-specific CAR can be expressed in T cells or NK cells using any of a variety of vectors, including lentiviral vectors and retroviral vectors.
  • pHIV7 vector developed at the T Cell Therapeutics Research Laboratory (TCTRL) at City of Hope (Duarte CA), is a useful vector for CAR expression. This vector uses the human EF1 promoter to drive expression of the CAR. Construction of pHIV7 is schematically depicted in FIGURE 1.
  • pv653RSN containing 653 bp from gag-pol plus 5’ and 3’ long-terminal repeats (LTRs) with an intervening SL3 -neomycin phosphotransferase gene (Neo), was subcloned into pBluescript, as follows: In Step 1, the sequences from 5’ LTR to rev-responsive element (RRE) made p5’HIV-l 51, and then the 5' LTR was modified by removing sequences upstream of the TATA box, and ligated first to a CMV enhancer and then to the SV40 origin of replication (p5'HIV-2).
  • RRE rev-responsive element
  • Step 2 after cloning the 3' LTR into pBluescript to make p3’HIV-1, a 400-bp deletion in the 3' LTR enhancer/promoter was made to remove cis-regulatory elements in HIV U3 and form p3'HIV-2.
  • Step 3 fragments isolated from the p5'HIV-3 and p3'HIV-2 were ligated to make pHIV-3.
  • Step 4 the p3'HIV-2 was further modified by removing extra upstream HIV sequences to generate p3 ’HIV-3 and a 600-bp BamHI-Sall fragment containing WPRE was added to p3 ’HIV-3 to make the p3HIV-4.
  • Step 5 the pHIV-3 RRE was reduced in size by PCR and ligated to a 5’ fragment from pHIV-3 (not shown) and to the p3’HIV-4, to make pHIV-6.
  • Step 6 a 190-bp Bglll-BamHI fragment containing the cPPT DNA flap sequence from HIV-1 pNL4-3 was amplified from pNL4-3 and placed between the RRE and the WPRE sequences in pHIV6 to make pHIV-7.
  • This parent plasmid pHIV7-GFP (GFP, green fluorescent protein) was used to package the parent vector using a four-plasmid system.
  • a packaging signal, psi y, is required for efficient packaging of viral genome into the vector.
  • the RRE and WPRE enhance the RNA transcript transport and expression of the transgene.
  • the flap sequence, in combination with WPRE, has been demonstrated to enhance the transduction efficiency of lentiviral vector in mammalian cells.
  • helper functions (which are required for production of the viral vector), are divided into three separate plasmids to reduce the probability of generation of replication competent lentivirus via recombination: 1) pCgp encodes the gag/pol protein required for viral vector assembly; 2) pCMV-Rev2 encodes the Rev protein, which acts on the RRE sequence to assist in the transportation of the viral genome for efficient packaging; and 3) pCMV-G encodes the glycoprotein of the vesiculo-stomatitis virus (VSV), which is required for infectivity of the viral vector.
  • VSV vesiculo-stomatitis virus
  • the regions of homology include a packaging signal region of approximately 600 nucleotides, located in the gag/pol sequence of the pCgp helper plasmid; a CMV promoter sequence in all three helper plasmids; and a RRE sequence in the helper plasmid pCgp. It is highly improbable that replication competent recombinant virus could be generated due to the homology in these regions, as it would require multiple recombination events. Additionally, any resulting recombinants would be missing the functional LTR and tat sequences required for lentiviral replication.
  • the CMV promoter was replaced by the EF1a-HTLV promoter (EF1p).
  • the EF1p has 563 bp and was introduced into epHIV7 using Nrul and Nhel, after the CMV promoter was excised.
  • the lentiviral genome, excluding gag/pol and rev that are necessary for the pathogenicity of the wild-type virus and are required for productive infection of target cells, has been removed from this system.
  • the vector construct does not contain an intact 3 ’LTR promoter, so the resulting expressed and reverse transcribed DNA proviral genome in targeted cells will have inactive LTRs.
  • compositions comprising, or alternatively consisting essentially of, or yet further consisting of, a carrier and any one or more of the polynucleotides as disclosed herein, a polypeptide as disclosed herein, any one of the vectors as disclosed herein, any one of the cells as disclosed herein, or a cell population as disclosed herein.
  • the carrier is a pharmaceutically acceptable carrier.
  • compositions of the present disclosure including but not limited to any one of the claimed compositions as described herein, in combination with one or more pharmaceutically or physiologically acceptable carriers, diluents or excipients.
  • Such compositions may comprise buffers such as neutral buffered saline, phosphate buffered saline and the like; carbohydrates such as glucose, mannose, sucrose or dextrans, mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; chelating agents such as EDTA or glutathione; adjuvants (e.g., aluminum hydroxide); and preservatives.
  • compositions of the present disclosure may be formulated for local or systemic administration, e.g., oral, intravenous, intracranial, topical, enteral, and/or parenteral administration. In certain embodiments, the compositions of the present disclosure are formulated for intravenous administration.
  • Administration of the cells or compositions can be effected in one dose, continuously or intermittently throughout the course of treatment and an effective amount to achieve the desired therapeutic benefit is provided.
  • Methods of determining the most effective means and dosage of administration are known to those of skill in the art and will vary with the composition used for therapy, the purpose of the therapy and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician. Suitable dosage formulations and methods of administering the agents are known in the art.
  • the cells and composition of the disclosure further comprises an agent used in a combined therapy.
  • the cells and composition of the disclosure can be administered in combination with other treatments or therapy.
  • a sample from the subject is isolated and assayed for FLT3 expression prior to administration of the disclosed therapy.
  • the methods further comprise assaying for FLT3 expression in a sample isolated from the subject, the sample containing a cancer or tumor cell.
  • the cells and populations of cell are administered to the host and/or subject using methods known in the art and described, for example, in PCT/US2011/064191.
  • this administration of the cells or compositions of the disclosure can be done for therapy or to generate an animal model of the desired disease, disorder, or condition for experimental and screening assays for the therapy or combination therapies
  • the disclosure is drawn to an isolated complex comprising, or alternatively consisting essentially of, or yet consisting of any one of the isolated cells as disclosed herein bound to a cancer or tumor cell, wherein the cancer or tumor cell is bound to the CAR-expressing isolated cell by the antigen binding domain of the antigen or tumor targeting antibody expressed by the CAR.
  • a method of treating a human patient suffering from cancer e.g. a FLT3 expressing cancer such as acute myeloid leukemia.
  • the method comprises, or alternatively consists essentially of, or yet further consists of administering a population of autologous or allogeneic human cells, e.g., immune cells, T or NK cells, stem cells, progenitor cells, cord blood cells, hematopoietic stem cells, comprising a nucleic acid molecule as disclosed herein.
  • the therapy can be combined with other therapies and can be administered as a first line, second line, third line, fourth line, or fifth line therapy.
  • the cancer can be a primary cancer or metastatic or recurrent cancer.
  • the patient has been treated with an agent to increase FLT3 expression on the cancer cell prior to being treated.
  • a method of treating a patient in need thereof is suffering from a FLT3 -expressing cancer or tumor.
  • the method comprises, or alternatively consists essentially of, or yet further consists of administering a cell population as disclosed herein to the patient.
  • the cells e.g., immune cells, T or NK cells, stem cells, progenitor cells, cord blood cells, hematopoietic stem cells can be allogenic or autologous to the patient being treated.
  • the therapy can be combined with other therapies and can be administered as a first line, second line, third line, fourth line, or fifth line therapy.
  • the cancer can be a primary cancer or metastatic or recurrent cancer.
  • the patient has been treated with an agent to increase FLT3 expression on the cancer cell prior to being treated.
  • the patient harbors tumor cell having an activating mutation in Fms-like tyrosine kinase 3 (FLT3). In further embodiments, the patient harbors tumor cell having an FLT3-ITD mutation.
  • FLT3 Fms-like tyrosine kinase 3
  • the chimeric antigen receptor expressing cell is administered locally or systemically.
  • the chimeric antigen receptor expressing cell is administered by single or repeat dosing. In some embodiments, the administration is repeated for at least once, or at least twice, or more times.
  • the cell population comprises, or alternatively consists essentially of, or yet further consists of an NK cell.
  • the NK cell is allogenic to the patient.
  • the method further comprises administering a second cell population as disclosed herein to the patient.
  • the second cell population comprises a CAR T cell.
  • the cells can be autologous or allogenic to the patient.
  • the CAR T cell is autologous to the patient.
  • the cell population comprises, or alternatively consists essentially of, or yet further consists of a T cell.
  • the method further comprises administering a drug increasing expression of FLT3 on a cancer cell.
  • the disclosure is drawn to a method of inhibiting the growth of a cancer cell expressing FLT3 or a tissue comprising the cancer cell.
  • the method comprises, or alternatively consists essentially of, or yet further consists of, contacting the cancer cell or tissue with any of the cells expressing the CARs and optionally the truncated protein maker as disclosed herein.
  • the contacting is ex vivo or in vitro or in vivo.
  • the contacting is in vivo and the expressing cells are autologous or allogeneic to a subject being treated.
  • the contacting is in vivo and the expressing cells are allogenic to a subject being treated.
  • the contacting in vivo can be by administration of the expressing cells to the subject in need of such treatment.
  • this disclosure provides a method of inhibiting the growth of a cancer cell expressing FLT3 or a tissue in a subject, comprising the cancer cell by administering to the subject, for example an effective amount of, the cells expressing the CAR as described herein and optionally a combined therapy.
  • the disclosure is drawn to a method of treating a cancer in a subject in need thereof.
  • the subject comprises a cancer cell expressing FLT3.
  • the method comprises, or alternatively consists essentially of, or yet further consists of, administering to the subject, for example an effective amount of, the cells expressing the CARs and optionally the truncated protein maker as disclosed herein.
  • the CAR expressing cells are autologous or allogeneic to a subject being treated.
  • the CAR expressing cells are allogenic to a subject being treated.
  • the method further comprises administering the subject a combined therapy.
  • the method comprises, or consists essentially of, or yet further consists of administering the cells expressing the CAR and optionally the truncated protein marker to the subject.
  • the CAR expressing cell is autologous or allogeneic to the subject in need.
  • the CAR expressing cell is allogenic to the subject in need.
  • a method for treating a cancer in a subject selected for the treatment comprises, or alternatively consists essentially of, or yet consists of administering, for example an effective amount of, a cell or a cell population as disclosed herein to the subject.
  • the subject is selected if a cancer cell of the subject expresses FLT3.
  • the FLT3 expression is determined by contacting a sample, such as a biopsy of a cancer, of the subject with an antibody or an antigen binding domain specifically recognizing and binding the FLT3 in vitro or in vivo, and detecting binding between the sample and the antibody or antigen binding domain.
  • the antigen binding domain further comprises a detectable marker.
  • the isolated or engineered cell is autologous to the subject in need. In some embodiments, the isolated or engineered cell is allogenic to the subject in need. [000263] In some aspects, a method as disclosed herein further comprises, or alternatively consists essentially of, or yet further consists of administering to the subject a combined therapy. Appropriate treatment regimens will be determined by the treating physician or veterinarian.
  • the FLT3 CAR cells may be administered before or after any one of these non-limiting exemplary combined therapies, e.g., before hematopoietic stem cell transplantation (HSCT) or after radiation therapy or chemotherapy.
  • HSCT hematopoietic stem cell transplantation
  • the FLT3 CAR cells may be used to achieve remission prior to the delivery of hematopoietic stem cells; in general, hematopoietic stem cell transplantation is more successful after remission.
  • non-limiting examples include other relevant cell types, such as unmodified immune cells, modified immune cells comprising vectors expressing one or more immunoregulatory molecules, or CAR cells specific to a different antigen than those disclosed herein. As with the CAR cells of the present disclosure, in some embodiments, these cells may be autologous or allogeneic.
  • the combined therapy is selected from one or both of a cytoreductive therapy or a therapy that upregulates the expression of FLT3 in a cancer cell.
  • the cytoreductive therapy comprises, or alternatively consists essentially of, or yet further consists of one or more of a chemotherapy, a cryotherapy, a hyperthermia, a targeted therapy, an immunotherapy, or a radiation therapy.
  • a combined therapy comprises, or alternatively consists essentially of, or yet further consists of physically removal of the cancer cells or tissue comprising the cancer cells.
  • the combined therapy may be used prior to, concurrently with, or after the FLT3 -specific CAR therapy as disclosed herein.
  • the therapy the upregulates the FLT3 expression in a cancer cell is used prior to or concurrently with the FLT3-specific CAR therapy as disclosed herein.
  • the immunotherapy refers to enhancing the immune response in the subject to a cancer cell, for example, an anti -PD- 1 therapy, or an anti-PD-Ll therapy.
  • a cancer cell for example, an anti -PD- 1 therapy, or an anti-PD-Ll therapy.
  • anti-PD- 1 therapy for example, an anti -PD- 1 therapy, or an anti-PD-Ll therapy.
  • commercially available antibodies to PD-1 include pembrolizumab (Merck), nivolumab (Bristol-Myers Squibb), pidilizumab (Cure Tech), AMP- 224 (GSK), AMP-514 (GSK), PDR001 (Novartis), and cemiplimab (Regeneron and Sanofi).
  • Non-limiting examples of commercially available antibodies to PD-L1 include atezolizumab (Roche Genentech), avelumab (Merck Soreno and Pfizer), durvalumab (AstraZeneca), BMS- 936559 (Bristol-Myers Suibb), and CK-301 (Checkpoint Therapeutics).
  • a method as disclosed herein is used as a first line therapy. In other embodiments, a method as disclosed herein is used as a second line therapy or a third line therapy.
  • the patient or subject maintains or recovers normal hematopoiesis after receiving, i.e., being administered, the effective amount of the isolated cell.
  • Normal hematopoiesis is a critical endpoint for certain cancers, such as but not limited to cancers affecting the blood or bone marrow, e.g., lymphoma or leukemia, such as but not limited to acute myeloid leukemia or acute lymphoblastic leukemia.
  • Methods of determining “normal hematopoiesis” after treatment are known in the art and include but are not limited to a “pin prick” blood test comparing baseline blood cell counts to post-treatment blood cell counts and/or similar comparisons for circulating CD34+ cells.
  • Non-limiting exemplary methods include bone marrow biopsy to verify engraftment. Failure to maintain or recover normal hematopoiesis (also known as normal engraftment) is associated with recurrent need for transfusions and/or need for antibiotics and/or high morbidity and mortality, in addition to symptomatic indicators such as but not limited to anemia, paleness, orthostatic hypotension, and bleeding and/or bruising due to a lack of platelet recovery.
  • symptomatic indicators such as but not limited to anemia, paleness, orthostatic hypotension, and bleeding and/or bruising due to a lack of platelet recovery.
  • Normal hematopoiesis and/or engraftment may be defined by a clinically acceptable threshold, such as but not limited to a sustained granulocyte count of > 1.0 x 10 9 /L, a sustained platelet count of > 50 x 10 9 , a sustained hemoglobin level of ⁇ 9 or 10 g/dL, and/or the absence of a need for red blood cell transfusions.
  • a clinically acceptable threshold such as but not limited to a sustained granulocyte count of > 1.0 x 10 9 /L, a sustained platelet count of > 50 x 10 9 , a sustained hemoglobin level of ⁇ 9 or 10 g/dL, and/or the absence of a need for red blood cell transfusions.
  • normal hematopoiesis is defined by a lack of significant depletion of Lin-CD34+CD38- CD90+CD45RA- cells.
  • adequate long-term hematopoiesis or successful long-term hematopoietic engraftment can be correlated with sufficient numbers of Lin-CD34+CD38-CD90+CD45RA- cells in the hematopoietic product being infused into a subject following myeloablative preparation for stem cell transplantation.
  • the total dose of CAR expressing cells may vary depending on, for example, the above disclosed factors.
  • the doses may be on the order of between 1 to 10 10 cells, e.g., at least 1, at least 10 1 , at least 10 2 , at least 10 3 , at least 10 4 , at least 10 5 , at least 10 6 , at least 10 7 , at most 10 8 , at most 10 9 , at most 10 10 , between 10 2 and 10 10 , between 10 3 and 10 9 , between 10 4 and 10 8 , per patient or per kilogram (kg) body weight of the patient.
  • the dose may be further limited by an integer coefficient to the order of magnitude, e.g., 1, 2, 3, 4, 5, 6, 7, 8, or 9, resulting a dose range listed according to the following non-limiting example: between 5 x 10 4 and 1 x 10 8 per patient or per kg body weight of the patient.
  • compositions of the present disclosure may be administered in a manner appropriate to the disease to be treated or prevented.
  • the quantity and frequency of administration will be determined by such factors as the condition of the patient, and the type and severity of the patient's disease, although appropriate dosages may be determined by clinical trials.
  • they are administered directly by direct injection or systemically such as intravenous injection.
  • aspects of the disclosure provide an exemplary method for determining if a patient is likely to respond to, or is not likely to respond to, CAR therapy.
  • the method comprises, or alternatively consists essentially of, or further consists of determining the presence or absence of necrosis in a tumor sample isolated from the patient and quantitating the amount of cancer or tumor cells expressing the cancer or tumor antigen.
  • the method further comprises, or alternatively consists essentially of, or yet further consists of administering an effective amount of the CAR therapy to the patient that is determined likely to respond to the CAR therapy.
  • the CAR therapy can be autologous or allogenic to the patient and the patient can be subject that suffers from a cancer, animal or human.
  • H&E staining hematoxylin and eosin stains
  • cytoplasmic H&E staining demonstrates increased eosinophilia, attributable in part to the loss of cytoplasmic RNA and in part to denatured cytoplasmic proteins.
  • necrotic tissue stains the cytoplasm often appears “moth eaten” due to enzyme digestion of cytoplasmic organelles.
  • necrotic tissues Myelin figures, calcification, and evidence of phagocytosis into other cells are also hallmarks of necrotic tissues that can be detected by histological staining. Necrotic tissues also have specific hallmarks in nuclear staining often demonstrating karyolysis, pyknosis, and karyorrhexis as a result of cell death. Using microscopy and either manual or automated quantitation of such necrotic hallmarks, relevance of CAR therapy may be determined. Alternate means of detecting tumorigenic or cancerous growth or necrotic tissues in general, including but not limited to biomarker-based or imaging-based diagnostics, are also equally relevant to determining whether a patient will respond to certain types of CAR therapy, and may be used accordingly. As is apparent, the CAR therapy is selected based on the genotype and/or phenotype of the cancer or tumor in the patient sample such that the antigen binding domain will target and treat the specific cancer or tumor.
  • kits that comprises, or alternatively consists essentially of, or yet further consists of, one or more of a CAR, a truncated protein marker, a polynucleotide, a cell, a cell population, or a composition as disclosed herein and optionally, instructions for making or using the same.
  • kits for performing these methods as well as instructions for carrying out the methods of the present disclosure such as collecting cells or tissues or both; performing the screening, transduction, etc.; analyzing the results, or any combination thereof.
  • the kit comprises, or alternatively consists essentially of, or yet further consists of, any one or more of: a polypeptide as disclosed herein, a polynucleotide as disclosed herein, a vector as disclosed herein, a vector comprising said nucleic acid, a cell as disclosed herein, such as isolated allogenic cells, preferably T cells or NK cells, a cell population as disclosed herein, a composition as disclosed herein, an isolated complex as disclosed herein, or instructions optionally on the procuring of autologous cells from a patient.
  • a kit may also comprise, or alternatively consist essentially of, or yet further comprise media and other reagents appropriate for the transduction, the selection, the activation, or the expansion of CAR.
  • the kit comprises, or alternatively consists essentially of, or yet further consists of, a CAR expressing cell or a population thereof.
  • the cells of this kit may require activation or expansion or both prior to administration to a subject in need thereof.
  • the kit may further comprise, or consist essentially of, media and reagents, such as those covered in the disclosure above, to activate or expand or both activate and expand the isolated CAR expressing cell.
  • the cell is to be used for a CAR therapy.
  • the kit comprises instructions on the administration of the isolated cell to a patient in need of CAR therapy.
  • kits of this disclosure can also comprise, e.g., a buffering agent, a preservative or a protein-stabilizing agent.
  • the kits can further comprise components necessary for detecting the detectable-label, e.g., an enzyme or a substrate.
  • the kits can also contain a control sample or a series of control samples, which can be assayed and compared to the test sample.
  • Each component of a kit can be enclosed within an individual container and all of the various containers can be within a single package, along with instructions for interpreting the results of the assays performed using the kit.
  • the kits of the present disclosure may contain a written product on or in the kit container. The written product describes how to use the reagents contained in the kit.
  • these suggested kit components may be packaged in a manner customary for use by those of skill in the art.
  • these suggested kit components may be provided in solution or as a liquid dispersion or the like.
  • Example 1 Vectors for expression of FLT3 CAR
  • FIGURE 2 A variety of vectors and expression systems can be used to transduce T cells to express a FLT3 CAR.
  • One suitable vector is depicted in FIGURE 2.
  • this lentiviral vector which is based on the pHIV7 vector (Chung et al. Mol Ther. 2014 May; 22(5): 952-963.
  • FLT3 CAR expression is driven by the EF-lalpha promoter and the FLT3 CAR is co-expressed with a truncated CD 19, which can be used as a marker.
  • FIGURE 3 An alternative vector for FLT3 CAR expression is depicted in FIGURE 3. Here expression is driven by an MMLV promoter and the FLT3 CAR is co-expressed with a truncated CD 19, which can be used as a marker.
  • PBMC enriched for CD4+ and CD8+ cells were activated using CD3/CD28 beads.
  • the vectors used the pHIV7 backbone.
  • the FLT3 -specific CAR were co-expressed with a CD19t tag, as described herein.
  • Two controls were used: a vector expressing GFP and a vector expressing the CD19t tag. In all cases the T cells were transfected at a MOI of 1. FLT3 CAR-T expression and cytotoxicity were performed on Day 5.
  • Tumor cells were obtained from 3 patients and were co-cultured with T cells transduced with the various constructs.
  • the expression of IL-2 (FIGURES 6A - 6C) and IFN-g (FIGURES 7A - 7C) in culture supernatants was measured.
  • FIGURES 4A - 4D and FIGURES 5A - 5B despite some interpatient variability, T cells transduced with the improved FLT3-specific CAR coding sequence were superior to T cells transduced with the earlier FLT3 -specific CAR coding sequence.
  • Example 5 FLT3 CAR activity in a mouse model of aggressive human FLT3(+) AML (MOLM-13).
  • Embodiment 1 A nucleic molecule comprising the nucleotide sequence of SEQ ID NO:l.
  • Embodiment 2 A viral vector or an expression vector comprising the nucleic acid molecule of Embodiment 1.
  • Embodiment 3 The nucleic acid molecule of Embodiment 1, further comprising a T2A skip sequence and a sequence encoding a truncated EGFR or a truncated CD 19.
  • Embodiment 4 The nucleic acid molecule of Embodiment 1, further comprising a T2A skip sequence and a sequence encoding a truncated EGFR.
  • Embodiment 5 The nucleic acid molecule of Embodiment 1, further comprising a T2A skip sequence and a sequence encoding a truncated CD 19.
  • Embodiment 6 The nucleic acid molecule of Embodiment 1, further comprising a T2A skip sequence and a sequence encoding a truncated EGFR.
  • Embodiment 7 The nucleic acid molecule of Embodiment 1, further comprising a T2A skip sequence and a sequence encoding a truncated CD 19.
  • Embodiment 8 The vector of Embodiment 2, wherein the vector is a lentiviral vector.
  • Embodiment 9 The vector of Embodiment 2, wherein the vector is a retroviral vector.
  • Embodiment 10 The vector of Embodiment 2, wherein expression of nucleotide sequence of SEQ ID NO: 1 is under the control of an EF-lalpha promoter or a MMLV promoter.
  • Embodiment 11 A population of human T cells transduced by a vector comprising the nucleic acid molecule of Embodiment 1.
  • Embodiment 12 The population of human T cells of Embodiment 11, wherein the population of human T cells comprise central memory T cells, NK cells, naive memory T cells, pan T cells, or PBMC substantially depleted for CD25+ cells and CD14+ cells.
  • Embodiment 13 A method of treating a patient suffering from acute myeloid leukemia, comprising administering a population of autologous or allogeneic human T cells transduced by a vector comprising the nucleic acid molecule of Embodiment 1.
  • Embodiment 14 The method of Embodiment 13, wherein the patient harbors tumor cell having an activating mutation in Fms-like tyrosine kinase 3 (FLT3).
  • FLT3 Fms-like tyrosine kinase 3
  • Embodiment 15 The method of Embodiment 13, wherein the patient harbors tumor cell having an FLT3-ITD mutation.
  • Embodiment 16 The method of Embodiment 13, wherein Embodiment chimeric antigen receptor is administered locally or systemically.
  • Embodiment 17 The method of Embodiment 13, wherein the chimeric antigen receptor is administered by single or repeat dosing.
  • Embodiment 18 A method of preparing CAR T cells comprising: providing a population of autologous or allogeneic human T cells and transducing the T cells by a vector comprising the nucleic acid molecule of Embodiment 1.
  • SEQ ID NO: 1 optimized CAR coding sequence
  • SEQ ID NO: 2 an FLT3 CAR
  • SEQ ID NO: 3 a signal sequence, MGW S SIILFL VATATGVH
  • SEQ ID NO: 4 an FLT3 scFv, wherein amino acid (aa) 1 to aa 118 of SEQ ID NO:
  • NO: 4 is a heavy chain variable region, aa 134 to aa 241 of SEQ ID NO: 4 is a light chain variable region, and aa 119 to aa 133 of SEQ ID NO: 4 is a peptide linker.
  • SEQ ID NO: 5 an IgGl hinge domain, LEPKSCDKTHTCPPCPDPKGT
  • SEQ ID NO: 6 a CD28 transmembrane domain
  • SEQ ID NO: 7 a CD28 costimulatory domain
  • SEQ ID NO: 8 a CD3zeta intracellular signaling domain
  • SEQ ID NO: 9 a ribosomal skip sequence
  • SEQ ID NO: 10 rEGFR
  • SEQ ID NO: 12 consensus sequence, wherein the small letter indicates any nucleotide or a nucleotide located in the corresponding position in SEQ ID NO: 1,
  • SEQ ID NO: 14 an scFv coding sequence prior to optimization
  • SEQ ID NO: 15 CDRLl, RASQSISNNLH [000321]
  • SEQ ID NO: 16 CDRL2, YASQSIS [000322]
  • SEQ ID NO: 17 CDRL3, QQSNTWPYT [000323]
  • SEQ ID NO: 18 CDRH1, SYWMH [000324]
  • SEQ ID NO: 19 CDRH2, EIDPSDSYKDYNQKFKD
  • SEQ ID NO : 20 CDRH3 , AITTTPFDF
  • SEQ ID NO: 21 an FLT3 heavy chain variable region
  • SEQ ID NO: 22 an FLT3 light chain variable region
  • SEQ ID NO : 24 MMLV promoter
  • SEQ ID NO: 25 FLT3 isoform 1
  • SEQ ID NO: 29 a 2 A peptide consensus motif

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Cell Biology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Oncology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Hematology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Developmental Biology & Embryology (AREA)
  • Virology (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Engineering & Computer Science (AREA)
EP21724442.5A 2020-04-17 2021-04-16 Gegen flt3 gerichtete chimäre antigenrezeptormodifizierte zellen zur behandlung von flt3-positiven malignomen Pending EP4135851A1 (de)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063011819P 2020-04-17 2020-04-17
PCT/US2021/027821 WO2021212069A1 (en) 2020-04-17 2021-04-16 Flt3-targeted chimeric antigen receptor modified cells for treatment of flt3-positive malignancies

Publications (1)

Publication Number Publication Date
EP4135851A1 true EP4135851A1 (de) 2023-02-22

Family

ID=75870743

Family Applications (1)

Application Number Title Priority Date Filing Date
EP21724442.5A Pending EP4135851A1 (de) 2020-04-17 2021-04-16 Gegen flt3 gerichtete chimäre antigenrezeptormodifizierte zellen zur behandlung von flt3-positiven malignomen

Country Status (11)

Country Link
US (1) US20230159644A1 (de)
EP (1) EP4135851A1 (de)
JP (1) JP2023522330A (de)
KR (1) KR20230004680A (de)
CN (1) CN115397517A (de)
AR (1) AR121880A1 (de)
AU (1) AU2021256053A1 (de)
CA (1) CA3175392A1 (de)
IL (1) IL297236A (de)
TW (1) TW202206452A (de)
WO (1) WO2021212069A1 (de)

Family Cites Families (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US5858358A (en) 1992-04-07 1999-01-12 The United States Of America As Represented By The Secretary Of The Navy Methods for selectively stimulating proliferation of T cells
US6534055B1 (en) 1988-11-23 2003-03-18 Genetics Institute, Inc. Methods for selectively stimulating proliferation of T cells
US6905680B2 (en) 1988-11-23 2005-06-14 Genetics Institute, Inc. Methods of treating HIV infected subjects
US6352694B1 (en) 1994-06-03 2002-03-05 Genetics Institute, Inc. Methods for inducing a population of T cells to proliferate using agents which recognize TCR/CD3 and ligands which stimulate an accessory molecule on the surface of the T cells
US7175843B2 (en) 1994-06-03 2007-02-13 Genetics Institute, Llc Methods for selectively stimulating proliferation of T cells
US5712149A (en) 1995-02-03 1998-01-27 Cell Genesys, Inc. Chimeric receptor molecules for delivery of co-stimulatory signals
US6692964B1 (en) 1995-05-04 2004-02-17 The United States Of America As Represented By The Secretary Of The Navy Methods for transfecting T cells
US7067318B2 (en) 1995-06-07 2006-06-27 The Regents Of The University Of Michigan Methods for transfecting T cells
US6846630B2 (en) 1996-10-18 2005-01-25 Takara Shuzo Co., Ltd. Nucleic acid encoding receptor type protein kinase
US6924123B2 (en) 1996-10-29 2005-08-02 Oxford Biomedica (Uk) Limited Lentiviral LTR-deleted vector
US6797514B2 (en) 2000-02-24 2004-09-28 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
MXPA02008265A (es) 2000-02-24 2004-09-10 Xcyte Therapies Inc Concentracion y estimulacion simultanea de calulas.
US6867041B2 (en) 2000-02-24 2005-03-15 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
US7419829B2 (en) 2000-10-06 2008-09-02 Oxford Biomedica (Uk) Limited Vector system
EP1967590A3 (de) 2001-03-13 2008-09-17 Novartis AG Lentivirale Verpackungskonstrukte
AR071891A1 (es) 2008-05-30 2010-07-21 Imclone Llc Anticuerpos humanos anti-flt3 (receptor tirosina cinasa 3 tipo fms humano)
CA2734128A1 (en) 2008-08-12 2010-02-18 Cellular Dynamics International, Inc. Methods for the production of ips cells
ES2791716T3 (es) 2010-12-14 2020-11-05 Univ Maryland Células T que expresan al receptor de antígeno quimérico antietiqueta universal y métodos para el tratamiento del cáncer
US9708384B2 (en) 2011-09-22 2017-07-18 The Trustees Of The University Of Pennsylvania Universal immune receptor expressed by T cells for the targeting of diverse and multiple antigens
CA2904014C (en) 2013-02-06 2021-06-15 Anthrogenesis Corporation Modified t lymphocytes having improved specificity
JP6450690B2 (ja) 2013-02-15 2019-01-09 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア キメラ抗原受容体及びその使用方法
KR102339240B1 (ko) 2013-10-15 2021-12-15 더 스크립스 리서치 인스티튜트 펩타이드 키메라 항원 수용체 t 세포 스위치 및 이의 용도
ES2741308T3 (es) 2013-10-15 2020-02-10 Scripps Research Inst Interruptores de células T con receptores de antígenos quiméricos y usos de los mismos
JP6560235B2 (ja) * 2014-01-13 2019-08-14 シティ・オブ・ホープCity of Hope Fcスペーサー領域に変異を有するキメラ抗原受容体(CAR)及びそれらの使用方法
CN104091269A (zh) 2014-06-30 2014-10-08 京东方科技集团股份有限公司 一种虚拟试衣方法及虚拟试衣系统
JP7113618B2 (ja) 2014-07-15 2022-08-05 ジュノー セラピューティクス インコーポレイテッド 養子細胞療法用の操作された細胞
AU2015338984A1 (en) 2014-10-31 2017-04-27 The Trustees Of The University Of Pennsylvania Methods and compositions for modified T cells
CA2966234A1 (en) 2014-12-15 2016-06-23 Bellicum Pharmaceuticals, Inc. Methods for controlled elimination of therapeutic cells
AU2015362753A1 (en) 2014-12-15 2017-04-27 Bellicum Pharmaceuticals, Inc. Methods for controlled activation or elimination of therapeutic cells
CN107614010A (zh) 2015-03-09 2018-01-19 艾更斯司股份有限公司 结合至flt3蛋白的抗体药物偶联物(adc)
TWI829617B (zh) 2015-07-31 2024-01-21 德商安美基研究(慕尼黑)公司 Flt3及cd3抗體構築體
EP3569244A1 (de) * 2015-09-23 2019-11-20 CytoImmune Therapeutics, LLC Flt3-gerichtete car-zellen zur immuntherapie
MY201207A (en) * 2016-04-01 2024-02-09 Amgen Inc Chimeric receptors to flt3 and methods of use thereof
PE20191846A1 (es) 2017-06-02 2019-12-31 Pfizer Anticuerpos especificos para flt3 y sus usos
SG10202108528QA (en) * 2017-06-02 2021-09-29 Pfizer Chimeric antigen receptors targeting flt3
JP7416695B2 (ja) * 2017-12-29 2024-01-17 メモリアル スローン-ケタリング キャンサー センター 増強されたキメラ抗原受容体およびその使用
WO2020010284A1 (en) 2018-07-04 2020-01-09 Cytoimmune Therapeutics, LLC Compositions and methods for immunotherapy targeting flt3, pd-1, and/or pd-l1
EP3623383A1 (de) * 2018-09-11 2020-03-18 Deutsches Krebsforschungszentrum, Stiftung des öffentlichen Rechts Verbesserte bispezifische flt3xcd3-antigenbindende proteine
WO2021050591A1 (en) * 2019-09-10 2021-03-18 Cytoimmune Therapeutics, Inc. Bispecific antibody car cell immunotherapy

Also Published As

Publication number Publication date
AR121880A1 (es) 2022-07-20
JP2023522330A (ja) 2023-05-30
IL297236A (en) 2022-12-01
KR20230004680A (ko) 2023-01-06
AU2021256053A1 (en) 2022-11-10
CA3175392A1 (en) 2021-10-21
US20230159644A1 (en) 2023-05-25
TW202206452A (zh) 2022-02-16
WO2021212069A1 (en) 2021-10-21
CN115397517A (zh) 2022-11-25

Similar Documents

Publication Publication Date Title
US20210269534A1 (en) Flt3 directed car cells for immunotherapy
ES2926513T3 (es) Métodos para evaluar la presencia o ausencia de virus competente en replicación
US20230348617A1 (en) Chimeric antigen receptors targeted to psca
US20230331872A1 (en) Bispecific antibody car cell immunotherapy
US20220281982A1 (en) Bispecific antibody car cell immunotherapy
JP2023540997A (ja) 免疫細胞を感染、活性化、および拡大させる方法および組成物
EP3841200A2 (de) Verfahren und zusammensetzungen zur genetischen modifikation von lymphozyten in blut oder in angereicherten pbmcs
US20230405122A1 (en) Compositions and uses of psca targeted chimeric antigen receptor modified cells
US20230159644A1 (en) Flt3-targeted chimeric antigen receptor modified cells for treatment of flt3-positive malignancies
WO2023230581A1 (en) Methods of manufacturing t cell therapies
Panchal Developing novel therapies for X-linked lymphoproliferative disease type 1
WO2024052318A1 (en) Novel dual split car-t cells for the treatment of cd38-positive hematological malignancies
AU2022229358A1 (en) Methods and compositions for the delivery of retroviral particles

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20221012

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)